1
|
Jaffery R, Zhao Y, Ahmed S, Schumacher JG, Ahn J, Shi L, Wang Y, Tan Y, Chen K, Tawbi H, Wang J, Schwarzschild MA, Peng W, Chen X. Soluble Immune Factor Profiles in Blood and CSF Associated with LRRK2 Mutations and Parkinson's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644460. [PMID: 40196608 PMCID: PMC11974741 DOI: 10.1101/2025.03.20.644460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background and Objectives Mutations in the Leucine-rich repeat kinase 2 (LRRK2) gene are one of the most common genetic causes of Parkinson's disease (PD) and are linked to immune dysregulation in both the central nervous system and periphery. However, peripheral and central profiles of soluble immune factors associated with LRRK2 mutations and PD have not been comprehensively characterized. Using serum and CSF samples from the LRRK2 Cohort Consortium (LCC), this study aimed to probe a broad range of soluble immune biomarkers associated with LRRK2 mutations and PD. Methods We investigated the levels of soluble immune regulators in the serum (n=651) and cerebrospinal fluid (CSF, n=129) of LRRK2 mutation carriers and non-carriers, both with and without PD. A total of 65 cytokines, chemokines, growth factors, and soluble receptors were assessed by Luminex immunoassay. A multivariable robust linear model was used to determine levels associated with LRRK2 mutations and PD status, adjusting for age, sex, and sample cohort. Correlations were assessed using the Spearman correlation coefficient. LRRK2 G2019S knock-in mice were used to validate the associations identified in the LCC. Results In this extensive discovery cohort, we identified several elevated serum immune regulatory factors associated with LRRK2 mutations. In particular, serum stromal cell-derived factor-1 alpha (SDF-1 alpha) levels, as supported by findings in LRRK2 G2019S knock-in mice, and tumor necrosis factor receptor II (TNF-RII) were significantly increased after multiple comparison adjustment. In contrast, LRRK2 mutations were associated with reduced soluble immune markers, including BAFF, CD40-Ligand, I-TAC, MIP-3 alpha, NGF beta, and IL-27 in CSF. Those with clinically diagnosed PD, with or without LRRK2 mutations, did not show strong signals in serum but reduced inflammatory analytes in CSF, including MIF, MMP-1, CD30, Tweak, and SDF-1 alpha. In addition, we found that the serum levels of these soluble immune factors display varied correlations with their corresponding CSF levels. Discussion This study highlights distinct immune profiles associated with LRRK2 mutations and PD in the periphery and CNS. Serum levels of SDF-1alpha and TNF-RII were elevated in LRRK2 mutation carriers, while CSF immune markers were reduced. In PD, irrespective of LRRK2 status, reduced CSF inflammatory analytes and weak serum signals were observed. These results provide insight into immune dysregulation linked to LRRK2 mutations. If replicable in independent datasets, they offer potential avenues for biomarker and therapeutic exploration.
Collapse
Affiliation(s)
- Roshni Jaffery
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Yuhang Zhao
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Sarfraz Ahmed
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Jackson G. Schumacher
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Jae Ahn
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Leilei Shi
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yujia Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yukun Tan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ken Chen
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hussein Tawbi
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian Wang
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael A. Schwarzschild
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Weiyi Peng
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Xiqun Chen
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
- Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| |
Collapse
|
2
|
He E, Li Y, Zhao R, Kong Q, Shao Y, Wang C, Liu B, Jiang Y, Liu Q, Cui H. IL7 as a Risk Factor for Prostate Cancer: Implications for T Cell Apoptosis and Infiltration in the Tumor Microenvironment. Prostate 2025; 85:315-323. [PMID: 39593187 DOI: 10.1002/pros.24830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/23/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Prostate cancer's complex interplay with the immune microenvironment prompted an investigation into immune-related pathogenic mechanisms and potential therapeutic targets. METHODS Within the GSE176031 data set, Seurat meticulously dissected single-cell profiles from radical prostatectomy patients. Leveraging CellMarker and SingleR cell identities were precisely annotated. Then, monocle traced pseudotime trajectories, illuminating cellular paths, complemented by CellChat's insights into intricate intercellular communications. Furthermore, mendelian randomization (MR) robustly substantiated causal associations within prostate cancer contexts. RESULTS Employing single-cell analysis on intraoperative tumor and normal tissue, we identified 15 distinct cell types, notably observing a significant T cell reduction in tumor samples. Intercellular communication analysis revealed multiple pathways between epithelial cells and T cells, highlighting interleukin (IL)-IL7R-IL2RG interactions. IL7R, crucial in T cell apoptosis, showed differential expression across T cell development stages. Patients with IL7 amplification had poorer outcomes (p < 0.05), supported by MR in two cohorts (ieu-b-4809 cohort: odds ratio [OR] = 1.005, p = 0.002, 95% confidence interval [CI] [1.002-1.008]; ebi-a-GCST90018905: OR = 1.063, p = 0.032, 95% CI [1.005-1.125]), confirming IL7 as a prostate cancer risk factor. CONCLUSIONS These findings suggest T cell depletion via IL7-IL7R signaling may drive prostate cancer progression, offering novel therapeutic insights.
Collapse
Affiliation(s)
- Enyang He
- Tianjin Medical University, Tianjin, China
- Graduate School of Tianjin Medical University, Tianjin, China
| | - Yaowen Li
- Tianjin Medical University, Tianjin, China
- The First Central Hospital of Tianjin, Tianjin, China
| | - Rui Zhao
- Tianjin Medical University, Tianjin, China
- General Hospital of Tianjin Medical University, Tianjin, China
| | - Qinyan Kong
- West China Hospital of Sichuan University, Chengdu, China
| | - Yi Shao
- Tianjin Medical University, Tianjin, China
- The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Cong Wang
- Tianjin Medical University, Tianjin, China
- General Hospital of Tianjin Medical University, Tianjin, China
| | - Baoqun Liu
- Tianjin Medical University, Tianjin, China
- The First Central Hospital of Tianjin, Tianjin, China
| | - Yvhang Jiang
- Tianjin Medical University, Tianjin, China
- General Hospital of Tianjin Medical University, Tianjin, China
| | - Qian Liu
- Tianjin Medical University, Tianjin, China
- The First Central Hospital of Tianjin, Tianjin, China
| | - Hualei Cui
- Tianjin Medical University, Tianjin, China
- Graduate School of Tianjin Medical University, Tianjin, China
| |
Collapse
|
3
|
Sweis RF, Chatta GS, Jain RK, Moon H, Delacroix SE, Fang A, D’Amico L, Kask AS, Cheever MA, Fling S, Sharon E, Lacroix A, Kaiser JC, Pachynski RK, Yu EY. A Phase II Open-Label, Randomized Clinical Trial of Atezolizumab with or without Human Recombinant IL-7 (CYT107) in Advanced Urothelial Cancer. Clin Cancer Res 2025; 31:299-307. [PMID: 39576210 PMCID: PMC11747792 DOI: 10.1158/1078-0432.ccr-24-1728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/08/2024] [Accepted: 11/19/2024] [Indexed: 01/18/2025]
Abstract
PURPOSE Advanced urothelial cancer generally has high mortality despite modern anti-PD-1/L1 antibody-based combinations. Augmenting checkpoint inhibitor-mediated immune responses with lymphocyte growth factors may improve outcomes. We conducted a randomized phase II study (Cancer Immunotherapy Trials Network-14) in 47 patients to explore whether human recombinant IL-7 (CYT107) could be safely combined with PD-L1 inhibition to enhance responses. PATIENTS AND METHODS Patients with urothelial cancer after platinum chemotherapy were randomized to atezolizumab alone or with CYT107 weekly for four doses. The primary objective was clinical efficacy by the objective response rate (ORR). Secondary objectives included safety, toxicity, and other clinical outcomes. Correlative endpoints included peripheral immunophenotyping and quantification of cytokines. RESULTS CYT107 plus atezolizumab was well-tolerated, without dose-limiting toxicities and lower grade 3 to 4 treatment-related adverse events compared with atezolizumab monotherapy. The ORR was 26.3% for the combination therapy versus 23.8% for atezolizumab alone (P = 0.428). The complete response rate was 10.5% for the combination therapy versus 4.8% for monotherapy. Three patients on combination therapy had responses >21 months versus one with monotherapy. CD4+ and CD8+ T-lymphocyte expansion occurred in patients with response to combination therapy, with the greatest effect in T memory stem cells. Patients who responded to treatment exhibited elevated baseline levels of CCL4 and reduced levels of VEGFA and TNF. CONCLUSIONS Combining CYT107 with atezolizumab was safe and resulted in lymphocyte expansion, a doubling of the complete response rate, and durable responses exceeding 2 years. However, the ORR was similar to atezolizumab alone. Increased and sustained doses of CYT107 coupled with patient selection strategies should be further investigated.
Collapse
Affiliation(s)
- Randy F. Sweis
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL
| | | | | | | | - Scott Edward Delacroix
- Louisiana State University School of Medicine and Stanley S. Scott Cancer Ctr, New Orleans, LA
| | | | | | | | | | | | | | | | | | | | - Evan Y. Yu
- Fred Hutchinson Cancer Center, Seattle, WA
- Division of Hematology and Oncology, Department of Medicine, University of Washington
| |
Collapse
|
4
|
Chuleerarux N, Makkoukdji N, Satnarine T, Kuhn JE, Nopsopon T, Valyasevi P, Schmidt FB, Kleiner G, Gans M. Inborn Errors of Immunity Presenting with Early-Onset Severe Atopy. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:62. [PMID: 39859044 PMCID: PMC11767231 DOI: 10.3390/medicina61010062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025]
Abstract
Inborn errors of immunity (IEIs), also known as primary immunodeficiencies, are a group of genetic disorders affecting the development and function of the immune system. While IEIs traditionally present with recurrent infections, an increasing number of cases manifest with early-onset severe atopy, including atopic dermatitis, food allergies, asthma, and allergic rhinitis-features that are often overlooked. This can lead to delayed diagnosis and treatment, which is crucial for IEI patients due to the risk of severe infections. We conducted a literature search and reviewed all IEIs that can present with early-onset severe atopy. The hallmark features of these disorders often include early-onset, persistent, and severe atopic dermatitis, food allergies, and recurrent episodes of asthma, which may be refractory to treatments. Additionally, we discuss the importance of recognizing such severe atopy as a potential indicator of an underlying immune deficiency, particularly when accompanied by unusual infections, growth failure, or autoimmunity. This review aims to raise awareness of this association and emphasize the need for early diagnosis and genetic testing in patients with atypical or treatment-resistant allergic diseases, allowing for more timely diagnosis of underlying immunodeficiencies and appropriate treatments.
Collapse
Affiliation(s)
- Nipat Chuleerarux
- Department of Internal Medicine, Jackson Memorial Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Nadia Makkoukdji
- Department of Pediatrics, Jackson Memorial Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Travis Satnarine
- Department of Pediatrics, Jackson Memorial Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jessica Elise Kuhn
- Department of Pediatrics, Jackson Memorial Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Tanawin Nopsopon
- Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Peerada Valyasevi
- Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Fernanda Bellodi Schmidt
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Gary Kleiner
- Division of Allergy/Immunology, Department of Pediatrics, Jackson Memorial Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Melissa Gans
- Division of Allergy/Immunology, Department of Pediatrics, Jackson Memorial Holtz Children’s Hospital, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
5
|
Islam MM, Watanabe E, Salma U, Ozaki M, Irahara T, Tanabe S, Katsuki R, Oishi D, Takeyama N. Immunoadjuvant therapy in the regulation of cell death in sepsis: recent advances and future directions. Front Immunol 2024; 15:1493214. [PMID: 39720718 PMCID: PMC11666431 DOI: 10.3389/fimmu.2024.1493214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 11/18/2024] [Indexed: 12/26/2024] Open
Abstract
Sepsis is characterized by a concomitant early pro-inflammatory response by immune cells to an infection, and an opposing anti-inflammatory response that results in protracted immunosuppression. The primary pathological event in sepsis is widespread programmed cell death, or cellular self-sacrifice, of innate and adaptive immune cells, leading to profound immunological suppression. This severe immune dysfunction hampers effective primary pathogen clearance, thereby increasing the risk of secondary opportunistic infections, latent viral reactivation, multiple organ dysfunction, and elevated mortality. The types of cell death include apoptosis (type I programmed cell death), autophagy (type II programmed cell death), NETosis (a program for formation of neutrophil extracellular traps (NETs)) and other programmed cell deaths like pyroptosis, ferroptosis, necroptosis, each contributing to immunosuppression in distinct ways during the later phases of sepsis. Extensive apoptosis of lymphocytes, such as CD4+, CD8+ T cells, and B cells, is strongly associated with immunosuppression. Apoptosis of dendritic cells further compromises T and B cell survival and can induce T cell anergy or promote regulatory Treg cell proliferation. Moreover, delayed apoptosis and impaired neutrophil function contribute to nosocomial infections and immune dysfunction in sepsis. Interestingly, aberrant NETosis and the subsequent depletion of mature neutrophils also trigger immunosuppression, and neutrophil pyroptosis can positively regulate NETosis. The interaction between programmed cell death 1 (PD-1) or programmed cell death 1 ligand (PD-L1) plays a key role in T cell modulation and neutrophil apoptosis in sepsis. The dendritic cell growth factor, Fms-like tyrosine kinase (FLTEL), increases DC numbers, enhances CD 28 expression, attenuates PD-L1, and improves survival in sepsis. Recently, immunoadjuvant therapies have attracted attention for their potential to restore host physiological immunity and homeostasis in patients with sepsis. This review focuses on several potential immunotherapeutic agents designed to bolster suppressed innate and adaptive immune responses in the management of sepsis.
Collapse
Affiliation(s)
- Md. Monirul Islam
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
- Department of Biochemistry and Biotechnology, University of Science and Technology Chittagong (USTC), Chattogram, Bangladesh
| | - Eizo Watanabe
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Umme Salma
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Masayuki Ozaki
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Takayuki Irahara
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Subaru Tanabe
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Ryusuke Katsuki
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Dai Oishi
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| | - Naoshi Takeyama
- Department of Emergency and Critical Care Medicine, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
6
|
Xu X, Chen H, Ren Z, Xu X, Wu W, Yang H, Wang J, Zhang Y, Zhou Q, Li H, Zhang S, Wang H, Xu C. Phase separation of chimeric antigen receptor promotes immunological synapse maturation and persistent cytotoxicity. Immunity 2024; 57:2755-2771.e8. [PMID: 39609126 DOI: 10.1016/j.immuni.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 09/25/2024] [Accepted: 11/06/2024] [Indexed: 11/30/2024]
Abstract
Major challenges of chimeric antigen receptor (CAR)-T cell therapy include poor antigen sensitivity and cell persistence. Here, we report a solution to these issues by exploiting CAR phase separation. We found that incorporation of an engineered T cell receptor CD3ε motif, EB6I, into the conventional 28Z or BBZ CAR induced self-phase separation through cation-π interactions. EB6I CAR formed a mature immunological synapse with the CD2 corolla to transduce efficient antigen and costimulatory signaling, although its tonic signaling remained low. Functionally, EB6I CAR-T cells exhibited improved signaling and cytotoxicity against low-antigen tumor cells and persistent tumor-killing function. In multiple primary and relapsed murine tumor models, EB6I CAR-T cells exerted better antitumor functions than conventional CAR-T cells against blood and solid cancers. This study thus unveils a CAR engineering strategy to improve CAR-T cell immunity by leveraging molecular condensation and signaling integration.
Collapse
Affiliation(s)
- Xinyi Xu
- Key Laboratory of Multi-cell Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haotian Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zhengxu Ren
- Key Laboratory of Multi-cell Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaomin Xu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Wei Wu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Haochen Yang
- Key Laboratory of Multi-cell Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - JinJiao Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yumeng Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Qiuping Zhou
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Hua Li
- Key Laboratory of Multi-cell Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shaoqing Zhang
- Key Laboratory of Multi-cell Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haopeng Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Chenqi Xu
- Key Laboratory of Multi-cell Systems, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
| |
Collapse
|
7
|
Zhang Y, Dong X, Zhang Y, Chen Z, Zhou G, Chen N, Shen W, Yang K, Pei P. Biomaterials to regulate tumor extracellular matrix in immunotherapy. J Control Release 2024; 376:149-166. [PMID: 39389365 DOI: 10.1016/j.jconrel.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
The tumor extracellular matrix (ECM) provides physical support and influences tumor development, metastasis, and the tumor microenvironment, creating barriers to immune drug delivery and cell infiltration. Therefore, modulating or degrading the ECM is of significant importance to enhance the efficacy of tumor immunotherapy. This manuscript initially summarizes the main strategies and mechanisms of biomaterials in modulating various components of the ECM, including collagen, fibronectin, hyaluronic acid, and in remodeling the ECM. Subsequently, it discusses the benefits of biomaterials for immunotherapy following ECM modulation, such as promoting the infiltration of drugs and immune cells, regulating immune cell function, and alleviating the immunosuppressive microenvironment. The manuscript also briefly introduces the application of biomaterials that utilize and mimic the ECM for tumor immunotherapy. Finally, it addresses the current challenges and future directions in this field, providing a comprehensive overview of the potential and innovation in leveraging biomaterials to enhance cancer treatment outcomes. Our work will offer a comprehensive overview of ECM modulation strategies and their application in biomaterials to enhance tumor immunotherapy.
Collapse
Affiliation(s)
- Yujie Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xuexue Dong
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Yanxiang Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zetong Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China
| | - Ni Chen
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| | - Wenhao Shen
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing Medical University, Jiangsu, China.
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pei Pei
- Department of Nuclear Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province 230022, China; Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, People's Republic of China.
| |
Collapse
|
8
|
Scapin G, Cagdas E, Grav LM, Lewis NE, Goletz S, Hafkenscheid L. Implications of glycosylation for the development of selected cytokines and their derivatives for medical use. Biotechnol Adv 2024; 77:108467. [PMID: 39447666 DOI: 10.1016/j.biotechadv.2024.108467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/13/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024]
Abstract
Cytokines are important regulators of immune responses, making them attractive targets for autoimmune diseases and cancer therapeutics. Yet, the significance of cytokine glycosylation remains underestimated. Many cytokines carry N- and O-glycans and some even undergo C-mannosylation. Recombinant cytokines produced in heterologous host cells may lack glycans or exhibit a different glycosylation pattern such as varying levels of galactosylation, sialylation, fucosylation or xylose addition compared to their human counterparts, potentially impacting critical immune interactions. We focused on cytokines that are currently utilized or designed in advanced therapeutic formats, including immunocytokines, fusokines, engager cytokines, and genetically engineered 'supercytokines.' Despite the innovative designs of these cytokine derivatives, their glycosylation patterns have not been extensively studied. By examining the glycosylation of the human native cytokines, G-CSF and GM-CSF, interferons β and γ, TNF-α and interleukins-2, -3 -4, -6, -7, -9, -12, -13, -15, -17A, -21, and - 22, we aim to assess its potential impact on their therapeutic derivatives. Understanding the glycosylation of the native cytokines could provide critical insights into the safety, efficacy, and functionality of these next-generation cytokine therapies, affecting factors such as stability, bioactivity, antigenicity, and half-life. This knowledge can guide the choice of optimal expression hosts for production and advance the development of effective cytokine-based therapeutics and synthetic immunology drugs.
Collapse
Affiliation(s)
- Giulia Scapin
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Ece Cagdas
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Lise Marie Grav
- Department of Biotechnology and Biomedicine, Mammalian Cell Line Engineering, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark; The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA; Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Steffen Goletz
- Department of Biotechnology and Biomedicine, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark.
| | - Lise Hafkenscheid
- Department of Biotechnology and Biomedicine, Biotherapeutic Glycoengineering and Immunology, Technical University of Denmark, Søltofts Plads, 2800 Kgs Lyngby, Denmark.
| |
Collapse
|
9
|
Desvaux E, Hemon P, Soret P, Le Dantec C, Chatzis L, Cornec D, Devauchelle-Pensec V, Elouej S, Duguet F, Laigle L, Poirier N, Moingeon P, Bretin S, Pers JO. High-content multimodal analysis supports the IL-7/IL-7 receptor axis as a relevant therapeutic target in primary Sjögren's syndrome. J Autoimmun 2024; 149:103147. [PMID: 38114349 DOI: 10.1016/j.jaut.2023.103147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 12/21/2023]
Abstract
OBJECTIVE While the involvement of IL-7/IL-7R axis in pSS has been described in relation to T cells, little is known about the contribution of this pathway in relationship with other immune cells, and its implication in autoimmunity. Using high-content multiomics data, we aimed at characterizing IL-7R expressing cells and the involvement of IL-7/IL-7R pathway in pSS pathophysiology. METHODS An IL-7 signature established using RNA-sequencing of human PBMCs incubated with IL-7 was applied to 304 pSS patients, and on RNA-Seq datasets from tissue biopsies. High-content immunophenotyping using flow and imaging mass cytometry was developed to characterize peripheral and in situ IL-7R expression. RESULTS We identified a blood 4-gene IL-7 module (IKZF4, KIAA0040, PGAP1 and SOS1) associated with anti-SSA/Ro positiveness in patients as well as disease activity, and a tissue 5-gene IL-7 module (IL7R, PCED1B, TNFSF8, ADAM19, MYBL1) associated with infiltration severity. We confirmed expression of IL-7R on T cells subsets, and further observed upregulation of IL-7R on double-negative (DN) B cells, and especially DN2 B cells. IL-7R expression was increased in pSS compared to sicca patients with variations seen according to the degree of infiltration. When expressed, IL-7R was mainly found on epithelial cells, CD4+ and CD8+ T cells, switched memory B cells, DN B cells and M1 macrophages. CONCLUSION This exhaustive characterization of the IL-7/IL-7R pathway in pSS pathophysiology established that two IL-7 gene modules discriminate pSS patients with a high IL-7 axis involvement. Their use could guide the implementation of an anti-IL-7R targeted therapy in a precision medicine approach.
Collapse
Affiliation(s)
- Emiko Desvaux
- LBAI, UMR1227, University of Brest, Inserm, Brest, France; Institut de Recherches Internationales Servier, Research and Development, Suresnes, France
| | - Patrice Hemon
- LBAI, UMR1227, University of Brest, Inserm, Brest, France
| | - Perrine Soret
- Institut de Recherches Internationales Servier, Research and Development, Suresnes, France
| | | | - Loukas Chatzis
- LBAI, UMR1227, University of Brest, Inserm, Brest, France; Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Divi Cornec
- LBAI, UMR1227, University of Brest, Inserm, Brest, France; CHU de Brest, Brest, France
| | | | - Sahar Elouej
- Institut de Recherches Internationales Servier, Research and Development, Suresnes, France
| | - Fanny Duguet
- Institut de Recherches Internationales Servier, Research and Development, Suresnes, France
| | - Laurence Laigle
- Institut de Recherches Internationales Servier, Research and Development, Suresnes, France
| | | | - Philippe Moingeon
- Institut de Recherches Internationales Servier, Research and Development, Suresnes, France
| | - Sylvie Bretin
- Institut de Recherches Internationales Servier, Research and Development, Suresnes, France
| | - Jacques-Olivier Pers
- LBAI, UMR1227, University of Brest, Inserm, Brest, France; CHU de Brest, Brest, France.
| |
Collapse
|
10
|
Miyamoto H, Kondo Y, Itobayashi E, Uehara M, Hiraoka A, Kudo M, Kakizaki S, Kagawa T, Miuma S, Suzuki T, Sugi K, Suyama K, Beppu T, Toyoda H, Yoshiji H, Uojima H, Miyase S, Inoue K, Tamori A, Ito T, Shimose S, Suda G, Hayashi T, Onishi M, Narahara S, Watanabe T, Iwatsuki M, Fukushima S, Tanaka Y. Evaluation of the associations of interlukin-7 genetic variants with toxicity and efficacy of immune checkpoint inhibitors: A replication study of a Japanese population, based on the findings of a European genome-wide association study. Hepatol Res 2024; 54:1215-1225. [PMID: 38990762 DOI: 10.1111/hepr.14092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/05/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
AIM Recent genome-wide association studies of European populations have identified rs16906115, a single-nucleotide polymorphism in the interleukin-7 gene, as a predictor of immune-related adverse events (irAEs) and the therapeutic efficacy of immune checkpoint inhibitors. We evaluated this single-nucleotide polymorphism in a Japanese population. METHODS From January 2021, we stored host DNA from individuals who received various types of immune checkpoint inhibitors. From this population, we categorized 510 participants into cases (grade ≥2 irAEs) and controls (received ≥3 immune checkpoint inhibitor doses, follow-up ≥12 weeks, no irAEs), and divided 339 hepatocellular carcinoma patients treated with atezolizumab/bevacizumab into responders and non-responders, evaluated using the modified response evaluation criteria in solid tumors. We compared the minor allele frequencies of rs16906115 between cases and controls, and responders and non-responders. RESULTS In the irAE prediction analysis of 234 cases and 276 controls, the minor allele frequency was 0.244 in the case group and 0.265 in the control group. This difference is not significant. In the analysis predicting the therapeutic efficacy for hepatocellular carcinoma patients, the responders had a significantly lower minor allele frequency of 0.220, compared with 0.300 for the non-responders (p = 0.022). Univariate and multivariate analyses identified the minor allele homozygosity as a significant predictor of treatment response, with odds ratios of 0.292 (p = 0.015) in the univariate analysis and 0.315 (p = 0.023) in the multivariate analysis. CONCLUSIONS In our Japanese cohort, no association was found between the rs16906115 minor allele and irAEs or treatment efficacy. The minor allele homozygosity may be associated with a negative therapeutic outcome. CLINICAL TRIAL REGISTRATION UMIN Clinical Trials Registry with the number UMIN000043798.
Collapse
Affiliation(s)
- Hideaki Miyamoto
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| | - Yasuteru Kondo
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
- Department of Hepatology, Sendai Tokushukai Hospital, Miyagi, Japan
- Department of Hepatology, Sendai Kousei Hospital, Miyagi, Japan
| | - Ei Itobayashi
- Department of Gastroenterology, Asahi General Hospital, Chiba, Japan
| | - Masayoshi Uehara
- Department of Gastroenterology, Saiseikai Kumamoto Hospital, Kumamoto, Japan
| | - Atsushi Hiraoka
- Department of Gastroenterology, Ehime Prefectural Central Hospital, Ehime, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Hospital, Osaka, Japan
| | - Satoru Kakizaki
- Department of Clinical Research, NHO Takasaki General Medical Center, Gunma, Japan
| | - Tatehiro Kagawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Kanagawa, Japan
| | - Satoshi Miuma
- Department of Gastroenterology and Hepatology, Nagasaki University Hospital, Nagasaki, Japan
| | - Takanori Suzuki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Science, Aichi, Japan
| | - Kazuhiro Sugi
- Department of Gastroenterology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Koichi Suyama
- Department of Medical Oncology, Toranomon Hospital, Tokyo, Japan
| | - Toru Beppu
- Department of Surgery, Yamaga City Medical Center, Kumamoto, Japan
| | - Hidenori Toyoda
- Department of Gastroenterology and Hepatology, Ogaki Municipal Hospital, Gifu, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology, Nara Medical University, Nara, Japan
| | - Haruki Uojima
- Department of Gastroenterology, Kitasato University Hospital, Kanagawa, Japan
| | - Shiho Miyase
- Department of Gastroenterology and Hepatology, Kumamoto Shinto General Hospital, Kumamoto, Japan
| | - Kaori Inoue
- Department of Liver and Diabetes and Endocrinology, Saga University Hospital, Saga, Japan
| | - Akihiro Tamori
- Department of Hepatology, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Hospital, Aichi, Japan
| | - Shigeo Shimose
- Department of Gastroenterology, Kurume University Hospital, Fukuoka, Japan
| | - Goki Suda
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Hokkaido, Japan
| | - Tsuguru Hayashi
- Department of Hepatology, Sendai Kousei Hospital, Miyagi, Japan
| | - Masaya Onishi
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
- Department of Gastroenterology, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Satoshi Narahara
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| | - Takehisa Watanabe
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Fukushima
- Faculty of Life Sciences, Department of Dermatology and Plastic Surgery, Kumamoto University, Kumamoto, Japan
| | - Yasuhito Tanaka
- Department of Gastroenterology and Hepatology, Kumamoto University Hospital, Kumamoto, Japan
| |
Collapse
|
11
|
Gao D, Yi XM, Feng L, Li S, Shu HB. MARCH8 Mediates K27-Linked Polyubiquitination of IL-7 Receptor α to Negatively Regulate IL-7-Triggered T Cell Homeostasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1467-1478. [PMID: 39311660 DOI: 10.4049/jimmunol.2400253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/09/2024] [Indexed: 11/06/2024]
Abstract
IL-7 is a cytokine produced by stromal cells, which binds to IL-7Rα and plays an important role for homeostasis of T lymphocytes. Excessive activities of IL-7-triggered signaling pathways causes autoimmune diseases. How IL-7-triggered signaling and immune effects are regulated is not fully understood. In this study, we show that the membrane-associated RING-CH (MARCH) E3 ligase family member MARCH8 mediates K27-linked polyubiquitination of IL-7Rα, leading to its lysosomal degradation. Site-directed mutagenesis suggests that MARCH8 meditates polyubiquitination of IL-7Rα at K265/K266, and mutation of these residues renders IL-7Rα resistance to MARCH8-mediated polyubiquitination and degradation. MARCH8 deficiency increases IL-7-triggered activation of the downstream transcription factor STAT5 and transcriptional induction of the effector genes in human T lymphoma cells. MARCH8 deficiency also promotes IL-7-triggered T cell proliferation and splenic memory CD8+ T cell differentiation in mice. Our findings suggest that MARCH8 negatively regulates IL-7-triggered signaling by mediating K27-linked polyubiquitination and lysosomal degradation of IL-7Rα, which reveals a negative regulatory mechanism of IL-7-triggered T cell homeostasis.
Collapse
Affiliation(s)
- Deng Gao
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China; and Research Unit of Innate Immune and Inflammatory Diseases (2019RU063), Chinese Academy of Medical Sciences, Wuhan, China
| | - Xue-Mei Yi
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China; and Research Unit of Innate Immune and Inflammatory Diseases (2019RU063), Chinese Academy of Medical Sciences, Wuhan, China
| | - Lu Feng
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China; and Research Unit of Innate Immune and Inflammatory Diseases (2019RU063), Chinese Academy of Medical Sciences, Wuhan, China
| | - Shu Li
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China; and Research Unit of Innate Immune and Inflammatory Diseases (2019RU063), Chinese Academy of Medical Sciences, Wuhan, China
| | - Hong-Bing Shu
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China; and Research Unit of Innate Immune and Inflammatory Diseases (2019RU063), Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
12
|
Dao HH, Nguyen TH, Hoang DH, Vu BD, Tran MA, Le MT, Hoang NTM, Bui AV, Than UTT, Nguyen XH. Manufacturing exosomes for wound healing: Comparative analysis of culture media. PLoS One 2024; 19:e0313697. [PMID: 39541412 PMCID: PMC11563385 DOI: 10.1371/journal.pone.0313697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Mesenchymal stem cell (MSC)-derived exosomes (EXs) have emerged as promising therapeutic agents for wound healing. However, the optimal conditions for manufacturing MSC-derived EXs that maximize their wound-healing potential have yet to be established. Hence, we compared the efficacy of five different MSC culture media, including three different serum-free, a platelet-supplemented, and a fetal bovine serum-supplemented media, in exosome manufacturing for wound healing applications. Although umbilical cord-derived MSCs (UCMSCs) cultured in these media exhibited similar proliferation, morphology, MSC surface marker expression, and stemness, EXs derived from UCMSCs cultured in different culture media displayed varying levels of growth factors and cytokines. Notably, EXs derived from platelet-supplemented media (DM-PLT_EXs) exhibited significantly higher concentrations of keratinocyte growth factor (KGF), vascular endothelial growth factor (VEGF-A), platelet-derived growth factor (PDGF-BB), interleukin 6 (IL-6), interleukin 7 (IL-7), and interleukin 8 (IL-8) than EXs from other media. These differences correlated with the superior capability of DM-PLT_EXs to promote human skin fibroblast proliferation and stimulate angiogenesis of human umbilical vein endothelial cells, making them a more suitable choice for wound healing applications. Our findings emphasize the significance of the culture medium selection in tailoring the therapeutic potential of UCMSC-derived EXs for wound healing.
Collapse
Affiliation(s)
- Huy Hoang Dao
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
- University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | | | | | - Bach Duong Vu
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Minh-Anh Tran
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Mai Thi Le
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
- Faculty of Biology, VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Nhung Thi My Hoang
- Faculty of Biology, VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Anh Viet Bui
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
| | - Uyen Thi Trang Than
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Xuan-Hung Nguyen
- Vinmec Hi-Tech Center, Vinmec Healthcare System, Hanoi, Vietnam
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
- College of Health Sciences, VinUniversity, Hanoi, Vietnam
| |
Collapse
|
13
|
Hui X, Farooq MA, Chen Y, Ajmal I, Ren Y, Xue M, Ji Y, Du B, Wu S, Jiang W. A novel strategy of co-expressing CXCR5 and IL-7 enhances CAR-T cell effectiveness in osteosarcoma. Front Immunol 2024; 15:1462076. [PMID: 39450160 PMCID: PMC11499113 DOI: 10.3389/fimmu.2024.1462076] [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: 07/17/2024] [Accepted: 09/17/2024] [Indexed: 10/26/2024] Open
Abstract
Background Solid tumors are characterized by a low blood supply, complex stromal architecture, and immunosuppressive milieu, which inhibit CAR-T cell entry and survival. CXCR5 has previously been employed to increase CAR-T cell infiltration into CXCL13+ cancers. On the other hand, IL-7 improves the survival and persistence of T cells inside a solid tumor milieu. Methods We constructed a novel NKG2D-based CAR (C5/IL7-CAR) that co-expressed CXCR5 and IL-7. The human osteosarcoma cell lines U-2 OS, 143B, and Mg63 highly expressed MICA/B and CXCL13, thus presenting a perfect avenue for the present study. Results Novel CAR-T cells are superior in their activation, degranulation, and cytokine release competence, hence lysing more target cells than conventional CAR. Furthermore, CXCR5 and IL-7 co-expression decreased the expression of PD-1, TIM-3, and TIGIT and increased Bcl-2 expression. Novel CAR-T cells show enhanced proliferation and differentiation towards the stem cell memory T cell phenotype. C5/IL7-CAR-T cells outperformed conventional CAR-T in eradicating osteosarcoma in mouse models and displayed better survival. Additionally, CXCR5 and IL-7 co-expression enhanced CAR-T cell numbers, cytokine release, and survival in implanted tumor tissues compared to conventional CAR-T cells. Mechanistically, C5/IL7-CAR-T cells displayed enhanced STAT5 signaling. Conclusion These findings highlight the potential of CXCR5 and IL-7 co-expression to improve CAR-T cell therapy efficacy against osteosarcoma.
Collapse
Affiliation(s)
- Xinhui Hui
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Muhammad Asad Farooq
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
- The First Dongguan Affiliated Hospital, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Yiran Chen
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Iqra Ajmal
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
- The First Dongguan Affiliated Hospital, Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Yaojun Ren
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
- College of Life Science, Xinjiang Normal University, Urumqi, China
| | - Min Xue
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yuzhou Ji
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Bingtan Du
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Shijia Wu
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| | - Wenzheng Jiang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
14
|
Yazdani R, Askari M, Ahmadi AM, Azizi G, Ciric B, Boehm A, Zhang GX, Rostami A. Mouse monocytes express CD127 by immune cells, not LPS. Front Immunol 2024; 15:1356004. [PMID: 39328413 PMCID: PMC11424446 DOI: 10.3389/fimmu.2024.1356004] [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: 12/14/2023] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
The essential role of interleukin 7 (IL-7) signaling via its receptor (IL-7Rα; CD127) in T cell development and function has been well documented. However, CD127 expression and function in myeloid cells, including monocytes, are less clear, especially in mice. In the present study we report an inducible CD127 expression in mouse monocytes/macrophages. This induction is dependent on the presence of other immune cells, highlighting that regulation of CD127 expression on monocytes differs in mice and humans. We demonstrate that CD127 is functional, as IL-7 downregulated its expression. We also saw decreased CD127 expression during inflammation in vivo. Overall, upregulation of CD127 expression in vitro and its downregulation in vivo confirm that CD127 is an inducible marker on mouse monocyte/macrophage cells, in contrast to findings recently published by others. Characterizing the role of CD127 signaling in myeloid cells in inflammatory disorders would be worthwhile in future study.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University,
Philadelphia, PA, United States
| |
Collapse
|
15
|
Li Z, Hu L, Wang Y, Liu Q, Liu J, Long H, Li Q, Luo L, Peng Y. Local administration of mRNA encoding cytokine cocktail confers potent anti-tumor immunity. Front Immunol 2024; 15:1455019. [PMID: 39290693 PMCID: PMC11406011 DOI: 10.3389/fimmu.2024.1455019] [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: 06/26/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Immunotherapy using inflammatory cytokines, such as interleukin (IL)-2 and interferon (IFN)-α, has been clinically validated in treating various cancers. However, systemic immunocytokine-based therapies are limited by the short half-life of recombinant proteins and severe dose-limiting toxicities. In this study, we exploited local immunotherapy by intratumoral administration of lipid nanoparticle (LNP)-encapsulated mRNA cocktail encoding cytokines IL-12, IL-7, and IFN-α. The cytokine mRNA cocktail induced tumor regression in multiple syngeneic mouse models and anti-tumor immune memory in one syngeneic mouse model. Additionally, immune checkpoint blockade further enhanced the anti-tumor efficacy of the cytokine mRNAs. Furthermore, human cytokine mRNAs exhibited robust anti-tumor efficacy in humanized mouse tumor models. Mechanistically, cytokine mRNAs induced tumor microenvironment inflammation, characterized by robust T cell infiltration and significant inflammatory cytokine and chemokine production.
Collapse
Affiliation(s)
- Zhigang Li
- Department of Research and Development, Liverna Therapeutics Inc., Zhuhai, China
| | - Ling Hu
- Department of Research and Development, Liverna Therapeutics Inc., Zhuhai, China
| | - Yi Wang
- Department of Research and Development, Liverna Therapeutics Inc., Zhuhai, China
| | - Qi Liu
- Department of Research and Development, Liverna Therapeutics Inc., Zhuhai, China
| | - Jun Liu
- Department of Research and Development, Liverna Therapeutics Inc., Zhuhai, China
| | - Haiyan Long
- Department of Research and Development, Liverna Therapeutics Inc., Zhuhai, China
| | - Qi Li
- Department of Research and Development, Liverna Therapeutics Inc., Zhuhai, China
| | - Liping Luo
- Department of Research and Development, Liverna Therapeutics Inc., Zhuhai, China
| | - Yucai Peng
- Department of Research and Development, Liverna Therapeutics Inc., Zhuhai, China
| |
Collapse
|
16
|
Di Nitto C, Ravazza D, Gilardoni E, Look T, Sun M, Prodi E, Moisoiu V, Pellegrino C, Manz MG, Puca E, Weller M, Weiss T, Neri D, De Luca R. An IL-7 fusion protein targeting EDA fibronectin upregulates TCF1 on CD8+ T-cells, preferentially accumulates to neoplastic lesions, and boosts PD-1 blockade. J Immunother Cancer 2024; 12:e008504. [PMID: 39142716 PMCID: PMC11332014 DOI: 10.1136/jitc-2023-008504] [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] [Accepted: 07/27/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Anti-PD-1 antibodies have revolutionized cancer immunotherapy due to their ability to induce long-lasting complete remissions in a proportion of patients. Current research efforts are attempting to identify biomarkers and suitable combination partners to predict or further improve the activity of immune checkpoint inhibitors. Antibody-cytokine fusions are a class of pharmaceuticals that showed the potential to boost the anticancer properties of other immunotherapies. Extradomain A-fibronectin (EDA-FN), which is expressed in most solid and hematological tumors but is virtually undetectable in healthy adult tissues, is an attractive target for the delivery of cytokine at the site of the disease. METHODS In this work, we describe the generation and characterization of a novel interleukin-7-based fusion protein targeting EDA-FN termed F8(scDb)-IL7. The product consists of the F8 antibody specific to the alternatively spliced EDA of FN in the single-chain diabody (scDb) format fused to human IL-7. RESULTS F8(scDb)-IL7 efficiently stimulates human peripheral blood mononuclear cells in vitro. Moreover, the product significantly increases the expression of T Cell Factor 1 (TCF-1) on CD8+T cells compared with an IL2-fusion protein. TCF-1 has emerged as a pivotal transcription factor that influences the durability and potency of immune responses against tumors. In preclinical cancer models, F8(scDb)-IL7 demonstrates potent single-agent activity and eradicates sarcoma lesions when combined with anti-PD-1. CONCLUSIONS Our results provide the rationale to explore the combination of F8(scDb)-IL7 with anti-PD-1 antibodies for the treatment of patients with cancer.
Collapse
Affiliation(s)
| | | | | | - Thomas Look
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
| | - Miaomiao Sun
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
| | | | - Vlad Moisoiu
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
| | - Christian Pellegrino
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Markus G. Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | - Michael Weller
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Dario Neri
- Philogen SpA, Siena, Italy
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
17
|
Zhao J, Zhang Y, Wang JY, Wei B, Liu YG. Expression pattern of serum interleukin-7 in elderly septic patients and its prognostic value for predicting short-term mortality. Cytokine 2024; 180:156664. [PMID: 38795605 DOI: 10.1016/j.cyto.2024.156664] [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: 02/19/2024] [Revised: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND The identification of novel prognostic biomarkers in elderly septic patients are essential for the improvement of mortality in sepsis in the context of precision medicine. The purpose of this study was to explore the expression pattern and prognostic value of serum interleukin-7 (IL-7) in predicting 28-day mortality in elderly patients with sepsis. METHODS Patients were retrospectively enrolled according to the sepsis-3.0 diagnostic criteria and divided into the survival group and non-survival group based on the clinical outcome at the 28-day interval. The baseline characteristic data, samples for the laboratory tests, and the SOFA, Acute Physiology and Chronic Health Evaluation (APACHE II), as well as Glasgow coma scale (GCS) scores, were recorded within 24 h after admission to the emergency department. Serum levels of IL-7 and TNF-α of the patients were quantified by the Luminex assay. Spearman correlation analysis, logistic regressive analysis and receiver operating characteristic curve (ROC) analysis were performed, respectively. RESULTS Totally, 220 elderly patients with sepsis were enrolled, 151 of whom died in a 28-day period. Albumin (ALB), high-density lipoprotein (HDL), systolic pressure (SBP), and platelet (PLT) were found to be significantly higher in the survival group (p < 0.05). IL-7 was shown to be correlated with TNF-α in the non-survival group (p = 0.030) but not in the survival group (p = 0.194). No correlation was shown between IL-7 and other factors (p > 0.05). IL-7 and TNF-α were found to be independent risk factors associated with the 28-day mortality (OR = 1.215, 1.420). Combination of IL-7, SOFA and ALB can make an AUROC of 0.874 with the specificity of 90.77 %. Combination of IL-7 and TNF-α can make an AUROC of 0.901 with the sensitivity of 90.41 % while the combination of IL-7, TNF-α, and ALB can make an AUROC of 0.898 with the sensitivity of 94.52 %. CONCLUSIONS This study highlights the importance of monitoring the serum level of IL-7 and TNF-α in elderly septic patients as well as evaluating the combinations with other routine risk factors which can be potentially used for the identification of elderly septic patients with higher risk of mortality.
Collapse
Affiliation(s)
- Jingjing Zhao
- Department of Infectious Disease and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China
| | - Ye Zhang
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Clinical Center for Medicine in Acute Infection, Capital Medical University, Beijing 100043, China
| | - Jun-Yu Wang
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Clinical Center for Medicine in Acute Infection, Capital Medical University, Beijing 100043, China
| | - Bing Wei
- Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Clinical Center for Medicine in Acute Infection, Capital Medical University, Beijing 100043, China.
| | - Yu-Geng Liu
- Department of Infectious Disease and Clinical Microbiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China; Emergency Medicine Clinical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China; Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Clinical Center for Medicine in Acute Infection, Capital Medical University, Beijing 100043, China.
| |
Collapse
|
18
|
Kim J, Maharjan R, Park J. Current Trends and Innovative Approaches in Cancer Immunotherapy. AAPS PharmSciTech 2024; 25:168. [PMID: 39044047 PMCID: PMC11573471 DOI: 10.1208/s12249-024-02883-x] [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: 01/30/2024] [Accepted: 07/05/2024] [Indexed: 07/25/2024] Open
Abstract
Immunotherapy is one of the most promising therapeutic approaches in the field of cancer treatment. As a tumor progresses, tumor cells employ an array of immune-regulatory mechanisms to suppress immune responses within the tumor microenvironment. Using our understanding of these mechanisms, cancer immunotherapy has been developed to enhance the immune system's effectiveness in treating cancer. Numerous cancer immunotherapies are currently in clinical use, yet many others are either in different stages of development or undergoing clinical studies. In this paper, we briefly discuss the features and current status of cancer immunotherapies. This includes the application of monoclonal antibodies, immune checkpoint inhibitors, adoptive cell therapy, cytokine therapy, cancer vaccines, and gene therapy, all of which have gained significant recognition in clinical practice. Additionally, we discuss limitations that may hinder successful clinical utilization and promising strategies, such as combining immunotherapy with nanotechnology.
Collapse
Affiliation(s)
- Jaechang Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| | - Ruby Maharjan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| | - Jonghyuck Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA.
- Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, USA.
- Markey Cancer Center, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
19
|
Costa JP, de Carvalho A, Paiva A, Borges O. Insights into Immune Exhaustion in Chronic Hepatitis B: A Review of Checkpoint Receptor Expression. Pharmaceuticals (Basel) 2024; 17:964. [PMID: 39065812 PMCID: PMC11279883 DOI: 10.3390/ph17070964] [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: 06/24/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Hepatitis B, caused by the hepatitis B virus (HBV), often progresses to chronic infection, leading to severe complications, such as cirrhosis, liver failure, and hepatocellular carcinoma. Chronic HBV infection is characterized by a complex interplay between the virus and the host immune system, resulting in immune cell exhaustion, a phenomenon commonly observed in chronic viral infections and cancer. This state of exhaustion involves elevated levels of inhibitory molecules, cells, and cell surface receptors, as opposed to stimulatory counterparts. This review aims to elucidate the expression patterns of various co-inhibitory and co-stimulatory receptors on immune cells isolated from chronic hepatitis B (CHB) patients. By analyzing existing data, the review conducts comparisons between CHB patients and healthy adults, explores the differences between HBV-specific and total T cells in CHB patients, and examines variations between intrahepatic and peripheral immune cells in CHB patients. Understanding the mechanisms underlying immune exhaustion in CHB is crucial for developing novel immunotherapeutic approaches. This detailed analysis sheds light on the immune exhaustion observed in CHB and lays the groundwork for future combined immunotherapy strategies aimed at leveraging checkpoint receptors to restore immune function and improve clinical outcomes.
Collapse
Affiliation(s)
- João Panão Costa
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal;
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Armando de Carvalho
- Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal; (A.d.C.); (A.P.)
- Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Artur Paiva
- Centro Hospitalar e Universitário de Coimbra, 3004-561 Coimbra, Portugal; (A.d.C.); (A.P.)
| | - Olga Borges
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal;
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
20
|
Jang JH, Choi E, Kim T, Yeo HJ, Jeon D, Kim YS, Cho WH. Navigating the Modern Landscape of Sepsis: Advances in Diagnosis and Treatment. Int J Mol Sci 2024; 25:7396. [PMID: 39000503 PMCID: PMC11242529 DOI: 10.3390/ijms25137396] [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: 05/31/2024] [Revised: 06/27/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Sepsis poses a significant threat to human health due to its high morbidity and mortality rates worldwide. Traditional diagnostic methods for identifying sepsis or its causative organisms are time-consuming and contribute to a high mortality rate. Biomarkers have been developed to overcome these limitations and are currently used for sepsis diagnosis, prognosis prediction, and treatment response assessment. Over the past few decades, more than 250 biomarkers have been identified, a few of which have been used in clinical decision-making. Consistent with the limitations of diagnosing sepsis, there is currently no specific treatment for sepsis. Currently, the general treatment for sepsis is conservative and includes timely antibiotic use and hemodynamic support. When planning sepsis-specific treatment, it is important to select the most suitable patient, considering the heterogeneous nature of sepsis. This comprehensive review summarizes current and evolving biomarkers and therapeutic approaches for sepsis.
Collapse
Affiliation(s)
- Jin Ho Jang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (J.H.J.); (E.C.); (T.K.); (H.J.Y.); (D.J.); (Y.S.K.)
- Department of Internal Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Eunjeong Choi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (J.H.J.); (E.C.); (T.K.); (H.J.Y.); (D.J.); (Y.S.K.)
- Department of Internal Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Taehwa Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (J.H.J.); (E.C.); (T.K.); (H.J.Y.); (D.J.); (Y.S.K.)
- Department of Internal Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hye Ju Yeo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (J.H.J.); (E.C.); (T.K.); (H.J.Y.); (D.J.); (Y.S.K.)
- Department of Internal Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Doosoo Jeon
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (J.H.J.); (E.C.); (T.K.); (H.J.Y.); (D.J.); (Y.S.K.)
- Department of Internal Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yun Seong Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (J.H.J.); (E.C.); (T.K.); (H.J.Y.); (D.J.); (Y.S.K.)
- Department of Internal Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Woo Hyun Cho
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (J.H.J.); (E.C.); (T.K.); (H.J.Y.); (D.J.); (Y.S.K.)
- Department of Internal Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
21
|
Romero-Mesones C, de Homdedeu M, Soler-Segovia D, Gómez-Ollés C, Espejo-Castellanos D, Ojanguren I, Saez-Gimenez B, Cruz MJ, Munoz X. The Impact of the Reduction in Environmental Pollution during COVID-19 Lockdown on Healthy Individuals. TOXICS 2024; 12:492. [PMID: 39058144 PMCID: PMC11280586 DOI: 10.3390/toxics12070492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024]
Abstract
The lockdown imposed to combat the COVID-19 pandemic produced a historic fall in air pollution in cities like Barcelona. This exceptional situation offered a unique context in which to examine the effects of air pollutants on human health. The present study aims to determine and compare the oxidative stress biomarkers Th1/Th2 and inflammatory-related cytokines in healthy individuals first during lockdown and then six months after the easing of the restrictions on mobility. A prospective study of a representative sample of 58 healthy, non-smoking adults was carried out. During lockdown and six months post-easing of restrictions, blood samples were drawn to measure the percentage of eosinophils, levels of Th1/Th2 and inflammatory-related cytokines assessed by a multiplex assay (BioRad Laboratories S.A., Marnes-la-Coquette, France), and levels of 8-isoprostane, glutathione peroxidase activity, and myeloperoxidase (Cayman Chemical Co., Ann Arbor, MI, USA), to assess their value as biomarkers of oxidative stress. Six months after easing mobility restrictions, increases in the levels of 8-isoprostane (p < 0.0001), IL-1β (p = 0.0013), IL-1ra (p = 0.0110), IL-4 (p < 0.0001), IL-13 (p < 0.0001), G-CSF (p = 0.0007), and CCL3 (p < 0.0001) were recorded, along with reductions in glutathione peroxidase (p < 0.0001), IFN-γ (p = 0.0145), TNFα (p < 0.0001), IP-10 (p < 0.0001), IL-2 (p < 0.0001), IL-7 (p < 0.0001), basic FGF (p < 0.0001), CCL4 (p < 0.0001), and CCL5 (p < 0.0001). No significant differences were observed in the rest of the biomarkers analyzed. The reduction in environmental pollution during the COVID-19 lockdown significantly lowered the levels of oxidative stress, systemic inflammation, and Th2-related cytokines in healthy people.
Collapse
Affiliation(s)
- Christian Romero-Mesones
- Pulmonology Service, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (C.R.-M.); (M.d.H.); (D.S.-S.); (C.G.-O.); (D.E.-C.); (I.O.); (B.S.-G.); (X.M.)
- CIBER Enfermedades Respiratorias (CibeRes), 28029 Madrid, Spain
- Medicine Department, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Miquel de Homdedeu
- Pulmonology Service, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (C.R.-M.); (M.d.H.); (D.S.-S.); (C.G.-O.); (D.E.-C.); (I.O.); (B.S.-G.); (X.M.)
- CIBER Enfermedades Respiratorias (CibeRes), 28029 Madrid, Spain
- Medicine Department, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - David Soler-Segovia
- Pulmonology Service, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (C.R.-M.); (M.d.H.); (D.S.-S.); (C.G.-O.); (D.E.-C.); (I.O.); (B.S.-G.); (X.M.)
- CIBER Enfermedades Respiratorias (CibeRes), 28029 Madrid, Spain
- Medicine Department, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Carlos Gómez-Ollés
- Pulmonology Service, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (C.R.-M.); (M.d.H.); (D.S.-S.); (C.G.-O.); (D.E.-C.); (I.O.); (B.S.-G.); (X.M.)
- CIBER Enfermedades Respiratorias (CibeRes), 28029 Madrid, Spain
- Medicine Department, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - David Espejo-Castellanos
- Pulmonology Service, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (C.R.-M.); (M.d.H.); (D.S.-S.); (C.G.-O.); (D.E.-C.); (I.O.); (B.S.-G.); (X.M.)
- CIBER Enfermedades Respiratorias (CibeRes), 28029 Madrid, Spain
- Medicine Department, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Inigo Ojanguren
- Pulmonology Service, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (C.R.-M.); (M.d.H.); (D.S.-S.); (C.G.-O.); (D.E.-C.); (I.O.); (B.S.-G.); (X.M.)
- CIBER Enfermedades Respiratorias (CibeRes), 28029 Madrid, Spain
- Medicine Department, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Berta Saez-Gimenez
- Pulmonology Service, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (C.R.-M.); (M.d.H.); (D.S.-S.); (C.G.-O.); (D.E.-C.); (I.O.); (B.S.-G.); (X.M.)
| | - María-Jesús Cruz
- Pulmonology Service, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (C.R.-M.); (M.d.H.); (D.S.-S.); (C.G.-O.); (D.E.-C.); (I.O.); (B.S.-G.); (X.M.)
- CIBER Enfermedades Respiratorias (CibeRes), 28029 Madrid, Spain
- Medicine Department, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Xavier Munoz
- Pulmonology Service, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain; (C.R.-M.); (M.d.H.); (D.S.-S.); (C.G.-O.); (D.E.-C.); (I.O.); (B.S.-G.); (X.M.)
- CIBER Enfermedades Respiratorias (CibeRes), 28029 Madrid, Spain
- Medicine Department, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| |
Collapse
|
22
|
Tachita T, Ri M, Aoki S, Asano A, Kanamori T, Totani H, Kinoshita S, Asao Y, Narita T, Masaki A, Ito A, Kusumoto S, Komatsu H, Iida S. Comprehensive analysis of serum cytokines in patients with multiple myeloma before and after lenalidomide and dexamethasone. Cancer Med 2024; 13:e70019. [PMID: 39031503 PMCID: PMC11259000 DOI: 10.1002/cam4.70019] [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: 01/31/2024] [Revised: 06/19/2024] [Accepted: 07/05/2024] [Indexed: 07/22/2024] Open
Abstract
Multiple myeloma (MM) is an incurable B-cell malignancy often accompanied by profound immunodeficiency. Lenalidomide (Len) is an immunomodulatory drug that exerts promising therapeutic effects on MM through the immune system. However, predictive markers related to the effects of Len treatment are not fully understood. This study aimed to identify candidate biomarkers for predicting the clinical efficacy of Len and dexamethasone (Ld) therapy through a comprehensive analysis of serum cytokines. The levels of 48 cytokines in the serum of patients with MM just before Ld therapy (n = 77), at the time of best response (n = 56), and at disease progression (n = 49) were measured and evaluated. Patients with high IL-18 and M-CSF levels showed significantly shorter progression-free survival and overall survival (OS). In contrast, patients with high PDGF-BB levels had longer survival. Moreover, low levels of G-CSF, IL-7, IL-8, and SDF-1α were associated with shorter OS after Ld therapy. During Ld therapy, pro-inflammatory cytokines such as IL-2Rα, IL-18, and TNF-α were decreased, while IFN-γ was increased. IL-4 and IL-6 levels increased during disease progression. In conclusion, this study provides a better understanding of the association between cytokines and the efficacy of Ld therapy as well as the unique changes in cytokines related to inflammatory and immune responses during Ld therapy.
Collapse
Affiliation(s)
- Takuto Tachita
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
- Department of Gastroenterology and HematologyHirosaki University Graduate School of MedicineHirosakiJapan
| | - Masaki Ri
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Sho Aoki
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Arisa Asano
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Takashi Kanamori
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Haruhito Totani
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Shiori Kinoshita
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Yu Asao
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Tomoko Narita
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Ayako Masaki
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Asahi Ito
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Shigeru Kusumoto
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Hirokazu Komatsu
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Shinsuke Iida
- Department of Hematology and OncologyNagoya City University Graduate School of Medical SciencesNagoyaJapan
| |
Collapse
|
23
|
Yang Y, Jeong J, Su T, Lai S, Zhang P, Garcia-Milian R, Graham M, Liu X, McConnell MJ, Utsumi T, Pereira J, Iwakiri Y. Interleukin-7-based identification of liver lymphatic endothelial cells reveals their unique structural features. JHEP Rep 2024; 6:101069. [PMID: 38966234 PMCID: PMC11222939 DOI: 10.1016/j.jhepr.2024.101069] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 07/06/2024] Open
Abstract
Background & Aims The lymphatic system plays crucial roles in maintaining fluid balance and immune regulation. Studying the liver lymphatics has been considered challenging, as common lymphatic endothelial cell (LyEC) markers are expressed by other liver cells. Additionally, isolation of sufficient numbers of LyECs has been challenging because of their extremely low abundance (<0.01% of entire liver cell population) in a normal liver. Methods Potential LyEC markers was identified using our published single-cell RNA sequencing (scRNA-seq) dataset (GSE147581) in mouse livers. Interleukin-7 (IL7) promoter-driven green fluorescent protein knock-in heterozygous mice were used for the validation of IL7 expression in LyECs in the liver, for the development of liver LyEC isolation protocol, and generating liver ischemia/reperfusion (I/R) injury. Scanning electron microscopy was used for the structural analysis of LyECs. Changes in LyEC phenotypes in livers of mice with I/R were determined by RNA-seq analysis. Results Through scRNA-seq analysis, we have identified IL7 as an exclusive marker for liver LyECs, with no overlap with other liver cell types. Based on IL7 expression in liver LyECs, we have established an LyEC isolation method and observed distinct cell surface structures of LyECs with fenestrae and cellular pores (ranging from 100 to 400 nm in diameter). Furthermore, we identified LyEC genes that undergo alterations during I/R liver injuries. Conclusions This study not only identified IL7 as an exclusively expressed gene in liver LyECs, but also enhanced our understanding of LyEC structures and demonstrated transcriptomic changes in injured livers. Impact and implications Understanding the lymphatic system in the liver is challenging because of the absence of specific markers for liver LyEC. This study has identified IL7 as a reliable marker for LyECs, enabling the development of an effective method for their isolation, elucidating their unique cell surface structure, and identifying LyEC genes that undergo changes during liver damage. The development of IL7 antibodies for detecting it in human liver specimens will further advance our understanding of the liver lymphatic system in the future.
Collapse
Affiliation(s)
- Yilin Yang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Jain Jeong
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Tingting Su
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
- Department of Gastroenterology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sanchuan Lai
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
- Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pengpeng Zhang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, China
| | - Rolando Garcia-Milian
- Bioinformatics Support Hub, Cushing/Whitney Medical Library, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Morven Graham
- Center for Cellular and Molecular Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Xinran Liu
- Center for Cellular and Molecular Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Matthew J. McConnell
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Teruo Utsumi
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Joao Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - Yasuko Iwakiri
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
24
|
Lee KJ, Choi D, Tae N, Song HW, Kang YW, Lee M, Moon D, Oh Y, Park S, Kim JH, Jeong S, Yang J, Park U, Hong DH, Byun MS, Park SH, Sohn J, Park Y, Im SK, Choi SS, Kim DH, Lee SW. IL-7-primed bystander CD8 tumor-infiltrating lymphocytes optimize the antitumor efficacy of T cell engager immunotherapy. Cell Rep Med 2024; 5:101567. [PMID: 38744277 PMCID: PMC11148861 DOI: 10.1016/j.xcrm.2024.101567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 02/29/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024]
Abstract
Bispecific T cell engagers (TCEs) show promising clinical efficacy in blood tumors, but their application to solid tumors remains challenging. Here, we show that Fc-fused IL-7 (rhIL-7-hyFc) changes the intratumoral CD8 T cell landscape, enhancing the efficacy of TCE immunotherapy. rhIL-7-hyFc induces a dramatic increase in CD8 tumor-infiltrating lymphocytes (TILs) in various solid tumors, but the majority of these cells are PD-1-negative tumor non-responsive bystander T cells. However, they are non-exhausted and central memory-phenotype CD8 T cells with high T cell receptor (TCR)-recall capacity that can be triggered by tumor antigen-specific TCEs to acquire tumoricidal activity. Single-cell transcriptome analysis reveals that rhIL-7-hyFc-induced bystander CD8 TILs transform into cycling transitional T cells by TCE redirection with decreased memory markers and increased cytotoxic molecules. Notably, TCE treatment has no major effect on tumor-reactive CD8 TILs. Our results suggest that rhIL-7-hyFc treatment promotes the antitumor efficacy of TCE immunotherapy by increasing TCE-sensitive bystander CD8 TILs in solid tumors.
Collapse
Affiliation(s)
- Kun-Joo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Donghoon Choi
- Research Institute of NeoImmuneTech, Inc., Pohang 37673, Republic of Korea
| | - Nara Tae
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Ha Won Song
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yeon-Woo Kang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Minji Lee
- Research Institute of NeoImmuneTech, Inc., Pohang 37673, Republic of Korea
| | - Dain Moon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Youngsik Oh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Sujeong Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Ji-Hae Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Siheon Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Jaehyuk Yang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Uni Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Da Hee Hong
- Genexine Inc., Seoul 07789, Republic of Korea
| | - Mi-Sun Byun
- Genexine Inc., Seoul 07789, Republic of Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon 34141, Republic of Korea
| | - Joohyuk Sohn
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yunji Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Sun-Kyoung Im
- Research Institute of NeoImmuneTech, Inc., Pohang 37673, Republic of Korea
| | - Sun Shim Choi
- Division of Biomedical Convergence, College of Biomedical Science, Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Dae Hee Kim
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon 24341, Republic of Korea; College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Republic of Korea.
| |
Collapse
|
25
|
Saeed MA, Peng B, Kim K, Rawat K, Kuehm LM, Siegel ZR, Borkowski A, Habib N, Van Tine B, Sheikh N, Tuyen V, Thorek DLJ, Fehniger TA, Pachynski RK. High-Dimensional Analyses Reveal IL15 Enhances Activation of Sipuleucel-T Lymphocyte Subsets and Reverses Immunoresistance. Cancer Immunol Res 2024; 12:559-574. [PMID: 38407894 DOI: 10.1158/2326-6066.cir-23-0652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/21/2023] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
Sipuleucel-T (sip-T) is the only FDA-approved autologous cellular immunotherapy for metastatic castration-resistant prostate cancer (mCRPC). To elucidate parameters of the response profile to this therapy, we report high-dimensional analyses of sip-T using cytometry by time of flight (CyTOF) and show a lymphoid predominance, with CD3+ T cells constituting the highest proportion (median ∼60%) of sip-T, followed by B cells, and natural killer (NK) and NKT cells. We hypothesized that treatment of sip-T with homeostatic cytokines known to activate/expand effector lymphocytes could augment efficacy against prostate tumors. Of the cytokines tested, IL15 was the most effective at enhancing activation and proliferation of effector lymphocytes, as well as augmenting tumor cytotoxicity in vitro. Co-culture of sip-T with IL15 and control or prostate-relevant antigens showed substantial activation and expansion of CD8+ T cells and NKT cells in an antigen-specific manner. Adoptive transfer of IL15-treated sip-T into NSG mice resulted in more potent prostate tumor growth inhibition compared with control sip-T. Evaluation of tumor-infiltrating lymphocytes revealed a 2- to 14-fold higher influx of sip-T and a significant increase in IFNγ producing CD8+ T cells and NKT cells within the tumor microenvironment in the IL15 group. In conclusion, we put forward evidence that IL15 treatment can enhance the functional antitumor immunity of sip-T, providing rationale for combining IL15 or IL15 agonists with sip-T to treat patients with mCRPC.
Collapse
Affiliation(s)
- Muhammad A Saeed
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Bo Peng
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Kevin Kim
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Kavita Rawat
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Lindsey M Kuehm
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Zoe R Siegel
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Ariel Borkowski
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Nabih Habib
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Brian Van Tine
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
| | | | - Vu Tuyen
- Dendreon Pharmaceuticals LLC, Seattle, Washington
| | - Daniel L J Thorek
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri
| | - Todd A Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
- Bursky Center for Human Immunology and Immunotherapy, Washington University School of Medicine, St Louis, Missouri
| | - Russell K Pachynski
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
- Bursky Center for Human Immunology and Immunotherapy, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
26
|
Eum HH, Jeong D, Kim N, Jo A, Na M, Kang H, Hong Y, Kong JS, Jeong GH, Yoo SA, Lee HO. Single-cell RNA sequencing reveals myeloid and T cell co-stimulation mediated by IL-7 anti-cancer immunotherapy. Br J Cancer 2024; 130:1388-1401. [PMID: 38424167 PMCID: PMC11014989 DOI: 10.1038/s41416-024-02617-7] [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: 06/08/2023] [Revised: 02/03/2024] [Accepted: 02/08/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors unleash inhibitory signals on T cells conferred by tumors and surrounding stromal cells. Despite the clinical efficacy of checkpoint inhibitors, the lack of target expression and persistence of immunosuppressive cells limit the pervasive effectiveness of the therapy. These limitations may be overcome by alternative approaches that co-stimulate T cells and the immune microenvironment. METHODS We analyzed single-cell RNA sequencing data from multiple human cancers and a mouse tumor transplant model to discover the pleiotropic expression of the Interleukin 7 (IL-7) receptor on T cells, macrophages, and dendritic cells. RESULTS Our experiment on the mouse model demonstrated that recombinant IL-7 therapy induces tumor regression, expansion of effector CD8 T cells, and pro-inflammatory activation of macrophages. Moreover, spatial transcriptomic data support immunostimulatory interactions between macrophages and T cells. CONCLUSION These results indicate that IL-7 therapy induces anti-tumor immunity by activating T cells and pro-inflammatory myeloid cells, which may have diverse therapeutic applicability.
Collapse
Affiliation(s)
- Hye Hyeon Eum
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Dasom Jeong
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Nayoung Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Areum Jo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Minsu Na
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Huiram Kang
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yourae Hong
- Digestive Oncology, Department of Oncology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jin-Sun Kong
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Gi Heon Jeong
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Seung-Ah Yoo
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Hae-Ock Lee
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
27
|
Li F, Dang W, Du Y, Xu X, He P, Zhou Y, Zhu B. Tuberculosis Vaccines and T Cell Immune Memory. Vaccines (Basel) 2024; 12:483. [PMID: 38793734 PMCID: PMC11125691 DOI: 10.3390/vaccines12050483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/27/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Tuberculosis (TB) remains a major infectious disease partly due to the lack of an effective vaccine. Therefore, developing new and more effective TB vaccines is crucial for controlling TB. Mycobacterium tuberculosis (M. tuberculosis) usually parasitizes in macrophages; therefore, cell-mediated immunity plays an important role. The maintenance of memory T cells following M. tuberculosis infection or vaccination is a hallmark of immune protection. This review analyzes the development of memory T cells during M. tuberculosis infection and vaccine immunization, especially on immune memory induced by BCG and subunit vaccines. Furthermore, the factors affecting the development of memory T cells are discussed in detail. The understanding of the development of memory T cells should contribute to designing more effective TB vaccines and optimizing vaccination strategies.
Collapse
Affiliation(s)
- Fei Li
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Wenrui Dang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Yunjie Du
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Xiaonan Xu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Pu He
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Yuhe Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
| | - Bingdong Zhu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Center for Tuberculosis Research, Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (F.L.); (W.D.); (Y.D.); (X.X.); (P.H.); (Y.Z.)
- College of Veterinary Medicine, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
28
|
Cao X, Fu YX, Peng H. Promising Cytokine Adjuvants for Enhancing Tuberculosis Vaccine Immunity. Vaccines (Basel) 2024; 12:477. [PMID: 38793728 PMCID: PMC11126114 DOI: 10.3390/vaccines12050477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis (M. tuberculosis), remains a formidable global health challenge, affecting a substantial portion of the world's population. The current tuberculosis vaccine, bacille Calmette-Guérin (BCG), offers limited protection against pulmonary tuberculosis in adults, underscoring the critical need for innovative vaccination strategies. Cytokines are pivotal in modulating immune responses and have been explored as potential adjuvants to enhance vaccine efficacy. The strategic inclusion of cytokines as adjuvants in tuberculosis vaccines holds significant promise for augmenting vaccine-induced immune responses and strengthening protection against M. tuberculosis. This review delves into promising cytokines, such as Type I interferons (IFNs), Type II IFN, interleukins such as IL-2, IL-7, IL-15, IL-12, and IL-21, alongside the use of a granulocyte-macrophage colony-stimulating factor (GM-CSF) as an adjuvant, which has shown effectiveness in boosting immune responses and enhancing vaccine efficacy in tuberculosis models.
Collapse
Affiliation(s)
- Xuezhi Cao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510182, China;
- Guangzhou National Laboratory, Bio-Island, Guangzhou 510005, China
| | - Yang-Xin Fu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Hua Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510182, China;
- Guangzhou National Laboratory, Bio-Island, Guangzhou 510005, China
| |
Collapse
|
29
|
Li X, Xu H, Du Z, Cao Q, Liu X. Advances in the study of tertiary lymphoid structures in the immunotherapy of breast cancer. Front Oncol 2024; 14:1382701. [PMID: 38628669 PMCID: PMC11018917 DOI: 10.3389/fonc.2024.1382701] [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: 02/13/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Breast cancer, as one of the most common malignancies in women, exhibits complex and heterogeneous pathological characteristics across different subtypes. Triple-negative breast cancer (TNBC) and HER2-positive breast cancer are two common and highly invasive subtypes within breast cancer. The stability of the breast microbiota is closely intertwined with the immune environment, and immunotherapy is a common approach for treating breast cancer.Tertiary lymphoid structures (TLSs), recently discovered immune cell aggregates surrounding breast cancer, resemble secondary lymphoid organs (SLOs) and are associated with the prognosis and survival of some breast cancer patients, offering new avenues for immunotherapy. Machine learning, as a form of artificial intelligence, has increasingly been used for detecting biomarkers and constructing tumor prognosis models. This article systematically reviews the latest research progress on TLSs in breast cancer and the application of machine learning in the detection of TLSs and the study of breast cancer prognosis. The insights provided contribute valuable perspectives for further exploring the biological differences among different subtypes of breast cancer and formulating personalized treatment strategies.
Collapse
Affiliation(s)
- Xin Li
- The First Clinical School of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Han Xu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ziwei Du
- The First Clinical School of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiang Cao
- Department of Earth Sciences, Kunming University of Science and Technology, Kunming, China
| | - Xiaofei Liu
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
30
|
Kim A, Oh MS, Lee GH, Song S, Byun MS, Choi D, Yu BY, Lee H. Understanding the pharmacokinetic journey of Fc-fusion protein, rhIL-7-hyFc using complementary approach of two analytical methods, accelerator mass spectrometry and ELISA. Antib Ther 2024; 7:105-113. [PMID: 38566969 PMCID: PMC10983079 DOI: 10.1093/abt/tbae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/21/2024] [Accepted: 01/28/2024] [Indexed: 04/04/2024] Open
Abstract
Antibody-based therapeutics (ABTs), including monoclonal/polyclonal antibodies and fragment crystallizable region (Fc)-fusion proteins, are increasingly used in disease treatment, driving the global market growth. Understanding the pharmacokinetic (PK) properties of ABTs is crucial for their clinical effectiveness. This study investigated the PK profile and tissue distribution of efineptakin alfa, a long-acting recombinant human interleukin-7 (rhIL-7-hyFc), using enzyme-linked immunosorbent assay (ELISA) and accelerator mass spectrometry (AMS). Totally, four rats were injected intramuscularly with 1 mg/kg of rhIL-7-hyFc containing 14C-rhIL-7-hyFc, which was prepared via reductive methylation. Serum total radioactivity (TRA) and serum rhIL-7-hyFc concentrations were quantified using AMS and ELISA, respectively. The TRA concentrations in organs were determined by AMS. Serum TRA peaked at 10 hours with a terminal half-life of 40 hours. The rhIL-7-hyFc exhibited a mean peak concentration at around 17 hours and a rapid elimination with a half-life of 12.3 hours. Peak concentration and area under the curve of TRA were higher than those of rhIL-7-hyFc. Tissue distribution analysis showed an elevated TRA concentrations in lymph nodes, kidneys, and spleen, indicating rhIL-7-hyFc's affinity for these organs. The study also simulated the positions of 14C labeling in rhIL-7-hyFc, identifying specific residues in the fragment of rhIL-7 portion, and provided the explanation of distinct analytes targeted by each method. Combining ELISA and AMS provided advantages by offering sensitivity and specificity for quantification as well as enabling the identification of analyte forms. The integrated use of ELISA and AMS offers valuable insights for the development and optimization of ABT.
Collapse
Affiliation(s)
- Anhye Kim
- Department of Clinical Pharmacology and Therapeutics, CHA Bundang Medical Center, CHA University, Seongnam 13496, Republic of Korea
- Department of Biomedical Informatics, CHA University School of Medicine, CHA University, Seongnam 13488, Republic of Korea
- Institute for Biomedical Informatics, CHA University School of Medicine, CHA University, Seongnam 13488, Republic of Korea
| | - Min-Seok Oh
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Gwan-Ho Lee
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Seongeun Song
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Republic of Korea
| | - Mi-sun Byun
- Clinical Development Division, Genexine, Inc., Seoul 07789, Republic of Korea
| | - Donghoon Choi
- Research Institute, NeoImmuneTech, co. Ltd., Pohang 37666, Republic of Korea
| | - Byung-Yong Yu
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Howard Lee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Advanced Institute of Convergence Technology, Suwon 16229, Republic of Korea
| |
Collapse
|
31
|
He J, Niu J, Wang L, Zhang W, He X, Zhang X, Hu W, Tang Y, Yang H, Sun J, Cui W, Shi Q. An injectable hydrogel microsphere-integrated training court to inspire tumor-infiltrating T lymphocyte potential. Biomaterials 2024; 306:122475. [PMID: 38306733 DOI: 10.1016/j.biomaterials.2024.122475] [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: 07/25/2023] [Revised: 12/14/2023] [Accepted: 01/17/2024] [Indexed: 02/04/2024]
Abstract
Although tumor-infiltrating T lymphocytes (TIL-Ts) play a crucial role in solid tumor immunotherapy, their clinical application has been limited because of the immunosuppressive microenvironment. Herein, we developed an injectable hydrogel microsphere-integrated training court (MS-ITC) to inspire the function of TIL-Ts and amplify TIL-Ts, through grafting with anti-CD3 and anti-CD28 antibodies and bovine serum albumin nanoparticles encapsulated with IL-7 and IL-15. MS-ITC provided the T-cell receptor and co-stimulatory signals required for TIL-Ts activation and IL-7/IL-15 signals for TIL-Ts expansion. Afterward, the MS-ITC was injected locally into the osteosarcoma tumor tissue in mice. MS-ITC suppressed the growth of primary osteosarcoma by more than 95 %, accompanied with primed and expanded TIL-Ts in the tumor tissues, compromising significantly increased CD8+ T and memory T cells, thereby enhancing the anti-tumor effect. Together, this work provides an injectable hydrogel microsphere-integrated training platform capable of inspiring TIL-Ts potential for a range of solid tumor immunotherapy.
Collapse
Affiliation(s)
- Jiachen He
- Department of Orthopedics, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Junjie Niu
- Department of Orthopedics, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Lin Wang
- Department of Orthopedics, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Wen Zhang
- Department of Orthopedics, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Xu He
- Department of Orthopedics, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Xiongjinfu Zhang
- Department of Orthopedics, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Wei Hu
- Department of Orthopedics, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Yunkai Tang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Huilin Yang
- Department of Orthopedics, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Jie Sun
- Department of Orthopedics, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China.
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China.
| | - Qin Shi
- Department of Orthopedics, National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China.
| |
Collapse
|
32
|
Tilsed CM, Sadiq BA, Papp TE, Areesawangkit P, Kimura K, Noguera-Ortega E, Scholler J, Cerda N, Aghajanian H, Bot A, Mui B, Tam Y, Weissman D, June CH, Albelda SM, Parhiz H. IL7 increases targeted lipid nanoparticle-mediated mRNA expression in T cells in vitro and in vivo by enhancing T cell protein translation. Proc Natl Acad Sci U S A 2024; 121:e2319856121. [PMID: 38513098 PMCID: PMC10990120 DOI: 10.1073/pnas.2319856121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/01/2024] [Indexed: 03/23/2024] Open
Abstract
The use of lipid nanoparticles (LNP) to encapsulate and deliver mRNA has become an important therapeutic advance. In addition to vaccines, LNP-mRNA can be used in many other applications. For example, targeting the LNP with anti-CD5 antibodies (CD5/tLNP) can allow for efficient delivery of mRNA payloads to T cells to express protein. As the percentage of protein expressing T cells induced by an intravenous injection of CD5/tLNP is relatively low (4-20%), our goal was to find ways to increase mRNA-induced translation efficiency. We showed that T cell activation using an anti-CD3 antibody improved protein expression after CD5/tLNP transfection in vitro but not in vivo. T cell health and activation can be increased with cytokines, therefore, using mCherry mRNA as a reporter, we found that culturing either mouse or human T cells with the cytokine IL7 significantly improved protein expression of delivered mRNA in both CD4+ and CD8+ T cells in vitro. By pre-treating mice with systemic IL7 followed by tLNP administration, we observed significantly increased mCherry protein expression by T cells in vivo. Transcriptomic analysis of mouse T cells treated with IL7 in vitro revealed enhanced genomic pathways associated with protein translation. Improved translational ability was demonstrated by showing increased levels of protein expression after electroporation with mCherry mRNA in T cells cultured in the presence of IL7, but not with IL2 or IL15. These data show that IL7 selectively increases protein translation in T cells, and this property can be used to improve expression of tLNP-delivered mRNA in vivo.
Collapse
Affiliation(s)
- Caitlin M. Tilsed
- Center for Cellular Immunology, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | | | - Tyler E. Papp
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Phurin Areesawangkit
- Center for Cellular Immunology, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok10700, Thailand
| | - Kenji Kimura
- Center for Cellular Immunology, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Estela Noguera-Ortega
- Center for Cellular Immunology, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - John Scholler
- Center for Cellular Immunology, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Nicholas Cerda
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | | | - Adrian Bot
- Capstan Therapeutics, San Diego, CA92121
| | - Barbara Mui
- Acuitas Therapeutics, Vancouver, BCV6T 1Z3, Canada
| | - Ying Tam
- Acuitas Therapeutics, Vancouver, BCV6T 1Z3, Canada
| | - Drew Weissman
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Carl H. June
- Center for Cellular Immunology, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Steven M. Albelda
- Center for Cellular Immunology, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Hamideh Parhiz
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
33
|
Fu C, Zhang X, Zhang X, Wang D, Han S, Ma Z. Advances in IL-7 Research on Tumour Therapy. Pharmaceuticals (Basel) 2024; 17:415. [PMID: 38675377 PMCID: PMC11054630 DOI: 10.3390/ph17040415] [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: 02/20/2024] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Interleukin-7 (IL-7) is a versatile cytokine that plays a crucial role in regulating the immune system's homeostasis. It is involved in the development, proliferation, and differentiation of B and T cells, as well as being essential for the differentiation and survival of naïve T cells and the production and maintenance of memory T cells. Given its potent biological functions, IL-7 is considered to have the potential to be widely used in the field of anti-tumour immunotherapy. Notably, IL-7 can improve the tumour microenvironment by promoting the development of Th17 cells, which can in turn promote the recruitment of effector T cells and NK cells. In addition, IL-7 can also down-regulate the expression of tumour growth factor-β and inhibit immunosuppression to promote anti-tumour efficacy, suggesting potential clinical applications for anti-tumour immunotherapy. This review aims to discuss the origin of IL-7 and its receptor IL-7R, its anti-tumour mechanism, and the recent advances in the application of IL-7 in tumour therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhenghai Ma
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China; (C.F.); (X.Z.); (X.Z.); (D.W.); (S.H.)
| |
Collapse
|
34
|
Lee M, Im SK, Baek S, Ji M, Kim M, Lee EJ, Ji ST, Ferrando-Martinez S, Wolfarth A, Lee JY, Kim D, Choi D. rhIL-7-hyFc and hIL-2/TCB2c combination promotes an immune-stimulatory tumor microenvironment that improves antitumor efficacy of checkpoint inhibitors. J Immunother Cancer 2024; 12:e008001. [PMID: 38471713 DOI: 10.1136/jitc-2023-008001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Recombinant human interleukin (rhIL)-7-hyFc (efineptakin alfa; NT-I7) is a potent T-cell amplifier, with two IL-7 molecules fused to IgD/IgG4 elements. rhIL-7-hyFc promotes extensive infiltration of CD8+ T cells into the tumor, concurrently increasing the numbers of intratumoral PD-1+CD8+ T cells. The hIL-2/TCB2 complex (SLC-3010) inhibits tumor growth by preferential activation of CD122 (IL-2Rβ)high CD8+ T cells and natural killer cells, over regulatory T cells (Tregs). We investigated the underlying mechanisms of rhIL-7-hyFc and hIL-2/TCB2c antitumor activity and the potential synergistic efficacy, specifically focusing on tumor-specific CD8+ cells within the tumor and the tumor-draining lymph nodes (tdLN). METHODS MC38 and CT26 tumor-bearing mice were administered with 10 mg/kg rhIL-7-hyFc intramuscularly and 0.9 mg/kg hIL-2/TCB2c intravenously. Anti-PD-1 monoclonal antibody was administered intraperitoneally three times at 3-day intervals at a dose of 5 mg/kg. Tumor volume was measured to assess efficacy. To compare the composition of immune cells between each monotherapy and the combination therapy, we analyzed tumors and tdLNs by flow cytometry. RESULTS Our data demonstrate that the combination of rhIL-7-hyFc and hIL-2/TCB2c increases efficacy and generates an immune-stimulatory tumor microenvironment (TME). The TME is characterized by an increased infiltration of tumor-specific CD8+ T cells, and a decreased frequency of CD39highTIM-3+ Treg cells. Most importantly, rhIL-7-hyFc increases infiltration of a CD62L+Ly108+ early progenitor population of exhausted CD8+ T cells (TPEX), which may retain long-term proliferation capacity and replenish functional effector CD8+ T cells. hIL-2/TCB2c induces differentiation of CD62L+Ly108+ TPEX rapidly into CD101+ terminally differentiated subsets (terminally exhausted T cell (TEX term)). Our study also demonstrates that rhIL-7-hyFc significantly enhances the proliferation rate of TPEX in the tdLNs, positively correlating with their abundance within the tumor. Moreover, rhIL-7-hyFc and hIL-2/TCB2c can overcome the limited therapeutic effectiveness of PD-1 blockade, culminating in the complete regression of tumors. CONCLUSIONS rhIL-7-hyFc can expand and maintain the progenitor pool of exhausted CD8+ T cells, whereas hIL-2/TCB2c promotes their differentiation into TEX term. Together, this induces an immune-stimulatory TME that improves the efficacy of checkpoint blockade.
Collapse
Affiliation(s)
- Minji Lee
- Research Institute of NeoImmuneTech, Co., Ltd, Pohang, Korea (the Republic of)
| | - Sun-Kyoung Im
- Research Institute of NeoImmuneTech, Co., Ltd, Pohang, Korea (the Republic of)
| | - Seungtae Baek
- Research Institute of NeoImmuneTech, Co., Ltd, Pohang, Korea (the Republic of)
| | - Mankyu Ji
- Research Institute of NeoImmuneTech, Co., Ltd, Pohang, Korea (the Republic of)
| | - Miyoung Kim
- Research Institute of NeoImmuneTech, Co., Ltd, Pohang, Korea (the Republic of)
| | - Eun Ju Lee
- Research Institute of NeoImmuneTech, Co., Ltd, Pohang, Korea (the Republic of)
| | - Seung Taek Ji
- Research Institute of NeoImmuneTech, Co., Ltd, Pohang, Korea (the Republic of)
| | | | | | | | - Daeun Kim
- Selecxine, Pohang, Korea (the Republic of)
| | - Donghoon Choi
- Research Institute of NeoImmuneTech, Co., Ltd, Pohang, Korea (the Republic of)
| |
Collapse
|
35
|
Kann MC, Schneider EM, Almazan AJ, Lane IC, Bouffard AA, Supper VM, Takei HN, Tepper A, Leick MB, Larson RC, Ebert BL, Maus MV, Jan M. Chemical genetic control of cytokine signaling in CAR-T cells using lenalidomide-controlled membrane-bound degradable IL-7. Leukemia 2024; 38:590-600. [PMID: 38123696 PMCID: PMC11774338 DOI: 10.1038/s41375-023-02113-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/19/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
CAR-T cell therapy has emerged as a breakthrough therapy for the treatment of relapsed and refractory hematologic malignancies. However, insufficient CAR-T cell expansion and persistence is a leading cause of treatment failure. Exogenous or transgenic cytokines have great potential to enhance CAR-T cell potency but pose the risk of exacerbating toxicities. Here we present a chemical-genetic system for spatiotemporal control of cytokine function gated by the off-patent anti-cancer molecular glue degrader drug lenalidomide and its analogs. When co-delivered with a CAR, a membrane-bound, lenalidomide-degradable IL-7 fusion protein enforced a clinically favorable T cell phenotype, enhanced antigen-dependent proliferative capacity, and enhanced in vivo tumor control. Furthermore, cyclical pharmacologic combined control of CAR and cytokine abundance enabled the deployment of highly active, IL-7-augmented CAR-T cells in a dual model of antitumor potency and T cell hyperproliferation.
Collapse
Affiliation(s)
- Michael C Kann
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Emily M Schneider
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA
| | - Antonio J Almazan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Isabel C Lane
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Amanda A Bouffard
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Valentina M Supper
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Hana N Takei
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Alexander Tepper
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mark B Leick
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Blood and Bone Marrow Transplant Program, Massachusetts General Hospital, Boston, MA, USA
| | - Rebecca C Larson
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Benjamin L Ebert
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Howard Hughes Medical Institute, Bethesda, MD, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Max Jan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
36
|
Dinges SS, Amini K, Notarangelo LD, Delmonte OM. Primary and secondary defects of the thymus. Immunol Rev 2024; 322:178-211. [PMID: 38228406 PMCID: PMC10950553 DOI: 10.1111/imr.13306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
The thymus is the primary site of T-cell development, enabling generation, and selection of a diverse repertoire of T cells that recognize non-self, whilst remaining tolerant to self- antigens. Severe congenital disorders of thymic development (athymia) can be fatal if left untreated due to infections, and thymic tissue implantation is the only cure. While newborn screening for severe combined immune deficiency has allowed improved detection at birth of congenital athymia, thymic disorders acquired later in life are still underrecognized and assessing the quality of thymic function in such conditions remains a challenge. The thymus is sensitive to injury elicited from a variety of endogenous and exogenous factors, and its self-renewal capacity decreases with age. Secondary and age-related forms of thymic dysfunction may lead to an increased risk of infections, malignancy, and autoimmunity. Promising results have been obtained in preclinical models and clinical trials upon administration of soluble factors promoting thymic regeneration, but to date no therapy is approved for clinical use. In this review we provide a background on thymus development, function, and age-related involution. We discuss disease mechanisms, diagnostic, and therapeutic approaches for primary and secondary thymic defects.
Collapse
Affiliation(s)
- Sarah S. Dinges
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kayla Amini
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
37
|
Cummings MJ, Bakamutumaho B, Lutwama JJ, Owor N, Che X, Astorkia M, Postler TS, Kayiwa J, Kiconco J, Muwanga M, Nsereko C, Rwamutwe E, Nayiga I, Kyebambe S, Haumba M, Bosa HK, Ocom F, Watyaba B, Kikaire B, Tomoiaga AS, Kisaka S, Kiwanuka N, Lipkin WI, O'Donnell MR. COVID-19 immune signatures in Uganda persist in HIV co-infection and diverge by pandemic phase. Nat Commun 2024; 15:1475. [PMID: 38368384 PMCID: PMC10874401 DOI: 10.1038/s41467-024-45204-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 01/17/2024] [Indexed: 02/19/2024] Open
Abstract
Little is known about the pathobiology of SARS-CoV-2 infection in sub-Saharan Africa, where severe COVID-19 fatality rates are among the highest in the world and the immunological landscape is unique. In a prospective cohort study of 306 adults encompassing the entire clinical spectrum of SARS-CoV-2 infection in Uganda, we profile the peripheral blood proteome and transcriptome to characterize the immunopathology of COVID-19 across multiple phases of the pandemic. Beyond the prognostic importance of myeloid cell-driven immune activation and lymphopenia, we show that multifaceted impairment of host protein synthesis and redox imbalance define core biological signatures of severe COVID-19, with central roles for IL-7, IL-15, and lymphotoxin-α in COVID-19 respiratory failure. While prognostic signatures are generally consistent in SARS-CoV-2/HIV-coinfection, type I interferon responses uniquely scale with COVID-19 severity in persons living with HIV. Throughout the pandemic, COVID-19 severity peaked during phases dominated by A.23/A.23.1 and Delta B.1.617.2/AY variants. Independent of clinical severity, Delta phase COVID-19 is distinguished by exaggerated pro-inflammatory myeloid cell and inflammasome activation, NK and CD8+ T cell depletion, and impaired host protein synthesis. Combining these analyses with a contemporary Ugandan cohort of adults hospitalized with influenza and other severe acute respiratory infections, we show that activation of epidermal and platelet-derived growth factor pathways are distinct features of COVID-19, deepening translational understanding of mechanisms potentially underlying SARS-CoV-2-associated pulmonary fibrosis. Collectively, our findings provide biological rationale for use of broad and targeted immunotherapies for severe COVID-19 in sub-Saharan Africa, illustrate the relevance of local viral and host factors to SARS-CoV-2 immunopathology, and highlight underemphasized yet therapeutically exploitable immune pathways driving COVID-19 severity.
Collapse
Affiliation(s)
- Matthew J Cummings
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA.
| | - Barnabas Bakamutumaho
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Julius J Lutwama
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Nicholas Owor
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Xiaoyu Che
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Maider Astorkia
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Thomas S Postler
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - John Kayiwa
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Jocelyn Kiconco
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | | | | | | | - Irene Nayiga
- Entebbe Regional Referral Hospital, Entebbe, Uganda
| | | | - Mercy Haumba
- Department of Arbovirology, Emerging and Re-emerging Infectious Diseases, Uganda Virus Research Institute, Entebbe, Uganda
| | - Henry Kyobe Bosa
- Uganda Peoples' Defence Forces, Kampala, Uganda
- Ministry of Health, Kampala, Uganda
| | | | - Benjamin Watyaba
- European and Developing Countries Clinical Trials Partnership-Eastern Africa Consortium for Clinical Research, Uganda Virus Research Institute, Entebbe, Uganda
| | - Bernard Kikaire
- European and Developing Countries Clinical Trials Partnership-Eastern Africa Consortium for Clinical Research, Uganda Virus Research Institute, Entebbe, Uganda
- Department of Pediatrics, Makerere University College of Health Sciences, Kampala, Uganda
| | - Alin S Tomoiaga
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Accounting, Business Analytics, Computer Information Systems, and Law, Manhattan College, New York, NY, USA
| | - Stevens Kisaka
- Department of Epidemiology and Biostatistics, Makerere University School of Public Health, Kampala, Uganda
- Institute of Tropical and Infectious Diseases, University of Nairobi, Nairobi, Kenya
| | - Noah Kiwanuka
- Department of Epidemiology and Biostatistics, Makerere University School of Public Health, Kampala, Uganda
| | - W Ian Lipkin
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Max R O'Donnell
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Center for Infection and Immunity, Mailman School of Public Health, Columbia University, New York, NY, USA
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, NY, USA
| |
Collapse
|
38
|
Kumar S, Bodhale N, Patra SK, Sarode A, Zhao L, Sarkar A, Saha B. Interleukin-7 potentiates MAPK10-elicited host-protective vaccine against Leishmania donovani. Cytokine 2024; 174:156475. [PMID: 38134556 DOI: 10.1016/j.cyto.2023.156475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/29/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023]
Abstract
Leishmania donovani causes the potentially fatal disease visceral leishmaniasis for which neither a vaccine nor an adjuvant for human use exists. Although interleukin-7 (IL-7) is implicated in CD4+ T-cell response stabilization, its anti-leishmanial function is uncertain. Therefore, we examined whether IL-7 would potentiate the efficacy of Leishmania major-expressed MAPK10 (LmjMAPK10; M10)-elicited anti-leishmanial host-protective response. We observed that aligning with IL-7R expression, IL-7 increased IFN-γ-secreting TH1 cell but reduced IL-4-producing TH2 cells and production of IL-10 and TGF-β effectuating anti-leishmanial functions in susceptible BALB/c mouse-derived macrophages. Co-culturing IL-7-pre-treated L. donovani-infected macrophages with L. donovani-infected BALB/c-derived T cells induced IFN-γ-dominated TH1 type anti-leishmanial function. IL-7 treatment of L. donovani-infected BALB/c mice significantly reduced splenic and hepatic parasite loads. Co-culturing CD4+ T cells from IL to 7-treated mice with L. donovani-infected macrophages reduced amastigote numbers suggesting IL-7-elicited host-protective effector T cells. Priming BALB/c with M10 + IL-7 reduced the splenic parasite burden more effectively than that was observed in M10-primed mice. An enhanced protection against L. donovani infection was accompanied by enhanced IL-12 and IFN-γ, but suppressed IL-10 and IL-4, response and host-protective TH1 and memory T cells. These results indicate IL-7-induced leishmanial antigen-specific memory T cell response that protects a susceptible host against L. donovani infection.
Collapse
Affiliation(s)
- Sunil Kumar
- National Centre for Cell Science, Ganeshkhind, Pune 411007. India
| | - Neelam Bodhale
- National Centre for Cell Science, Ganeshkhind, Pune 411007. India
| | | | - Aditya Sarode
- National Centre for Cell Science, Ganeshkhind, Pune 411007. India
| | - Ling Zhao
- Ling Zhao, Huazhong Agricultural University, Wuhan 430070, China
| | - Arup Sarkar
- Trident Academy of Creative Technology, Bhubaneswar 751024. India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411007. India; Trident Academy of Creative Technology, Bhubaneswar 751024. India.
| |
Collapse
|
39
|
Liu Y, Dang Y, Zhang C, Liu L, Cai W, Li L, Fang L, Wang M, Xu S, Wang G, Zheng J, Li H. IL-21-armored B7H3 CAR-iNKT cells exert potent antitumor effects. iScience 2024; 27:108597. [PMID: 38179061 PMCID: PMC10765065 DOI: 10.1016/j.isci.2023.108597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 10/06/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024] Open
Abstract
CD1d-restricted invariant NKT (iNKT) cells play a critical role in tumor immunity. However, the scarcity and limited persistence restricts their development and clinical application. Here, we demonstrated that iNKT cells could be efficiently expanded using modified cytokines combination from peripheral blood mononuclear cells. Introduction of IL-21 significantly increased the frequency of CD62L-positive memory-like iNKT cells. iNKT cells armoring with B7H3-targeting second generation CAR and IL-21 showed potent tumor cell killing activity. Moreover, co-expression of IL-21 promoted the activation of Stat3 signaling and reduced the expression of exhaustion markers in CAR-iNKT cells in vitro. Most importantly, IL-21-arming significantly prolonged B7H3 CAR-iNKT cell proliferation and survival in vivo, thus improving their therapeutic efficacy in mouse renal cancer xerograph models without observed cytokine-related adverse events. In summary, these results suggest that B7H3 CAR-iNKT armored with IL-21 is a promising therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Yilin Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Yuanyuan Dang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Chuhan Zhang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Liu Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Wenhui Cai
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Liantao Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Med-ical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Med-ical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Meng Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Med-ical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Shunzhe Xu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Med-ical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Junnian Zheng
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Med-ical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Med-ical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| |
Collapse
|
40
|
Tomala J, Cao SD, Spangler JB. Engineering Anticytokine Antibodies for Immune Modulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:225-234. [PMID: 38166248 DOI: 10.4049/jimmunol.2300467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/28/2023] [Indexed: 01/04/2024]
Abstract
The delicate balance of immune homeostasis is regulated by the interactions between cytokines and their cognate cell surface signaling receptors. There is intensive interest in harnessing cytokines as drugs for diseases such as cancer and autoimmune disorders. However, the multifarious and often contradictory activities of cytokines, coupled with their short serum half-lives, limit clinical performance and result in dangerous toxicities. There is thus growing emphasis on manipulating natural cytokines to enhance their selectivity, safety, and durability through various strategies. One strategy that has gained traction in recent years is the development of anticytokine Abs that not only extend the circulation half-life of cytokines but also specifically bias their immune activities through multilayered molecular mechanisms. Although Abs are notorious for their antagonistic activities, this review focuses on anticytokine Abs that selectively agonize the activity of the target protein. This approach has potential to help realize the clinical promise of cytokine-based therapies.
Collapse
Affiliation(s)
- Jakub Tomala
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University School of Engineering, Baltimore, MD
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Shanelle D Cao
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University School of Engineering, Baltimore, MD
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jamie B Spangler
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University School of Engineering, Baltimore, MD
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| |
Collapse
|
41
|
Kong Y, Wang X, Xu H, Liu S, Qie R. A Mendelian randomization study on the causal association of circulating cytokines with colorectal cancer. PLoS One 2023; 18:e0296017. [PMID: 38096329 PMCID: PMC10721084 DOI: 10.1371/journal.pone.0296017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Circulating cytokines have been associated with colorectal cancer (CRC). However, their causal correlation remains undetermined. This investigation uses genetic data to evaluate the mechanism that links circulating cytokines and CRC via Mendelian Randomization (MR). METHODS A two-sample MR evaluation was carried out to investigate the mechanism associating circulating cytokines and CRC in individuals of European ancestry. The Genome-wide association studies statistics, which are publically accessible, were used. Eligible instrumental SNPs that were significantly related to the circulating cytokines were selected. Multiple MR analysis approaches were carried out, including Simple Mode, inverse variance weighted (IVW), MR-Egger, Weighted Mode, Weighted Median, and MR pleiotropy residual sum and outlier (MR-PRESSO) methods. RESULTS The evidence supporting the association of genetically predicted circulating levels with the increased risk of CRC was revealed; these included vascular endothelial growth factor (OR = 1.352, 95% CI: 1.019-1.315, P = 0.024), interleukin-12p70 (OR = 1.273, 95% CI: 1.133-1.430, P = 4.68×10-5), interleukin-13 (OR = 1.149, 95% CI: 1.012-1.299, P = 0.028), interleukin-10 (OR = 1.230, 95% CI: 1.013-1.493, P = 0.037), and interleukin-7 (OR = 1.191, 95% CI: 1.023-1.386 P = 0.024). Additionally, MR analysis negative causal association between macrophage colony stimulating factor and CRC (OR = 0.854, 95% CI: 0.764-0.955, P = 0.005). The data from Simple Mode, Weighted Median, MR-Egger, and Weighted Mode analyses were consistent with the IVW estimates. Furthermore, the sensitivity analysis indicated that the presence of no horizontal pleiotropy to bias the causal estimates. CONCLUSION This investigation identified a causal association between circulating cytokines levels risk of CRC and may provide a deeper understanding of the pathogenesis of CRC, as well as offer promising leads for the development of novel therapeutic targets for CRC.
Collapse
Affiliation(s)
- Youqian Kong
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaoyu Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Hongyun Xu
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shaoxuan Liu
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Rui Qie
- Department of Internal Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
42
|
Luo M, Gong W, Zhang Y, Li H, Ma D, Wu K, Gao Q, Fang Y. New insights into the stemness of adoptively transferred T cells by γc family cytokines. Cell Commun Signal 2023; 21:347. [PMID: 38049832 PMCID: PMC10694921 DOI: 10.1186/s12964-023-01354-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/11/2023] [Indexed: 12/06/2023] Open
Abstract
T cell-based adoptive cell therapy (ACT) has exhibited excellent antitumoral efficacy exemplified by the clinical breakthrough of chimeric antigen receptor therapy (CAR-T) in hematologic malignancies. It relies on the pool of functional T cells to retain the developmental potential to serially kill targeted cells. However, failure in the continuous supply and persistence of functional T cells has been recognized as a critical barrier to sustainable responses. Conferring stemness on infused T cells, yielding stem cell-like memory T cells (TSCM) characterized by constant self-renewal and multilineage differentiation similar to pluripotent stem cells, is indeed necessary and promising for enhancing T cell function and sustaining antitumor immunity. Therefore, it is crucial to identify TSCM cell induction regulators and acquire more TSCM cells as resource cells during production and after infusion to improve antitumoral efficacy. Recently, four common cytokine receptor γ chain (γc) family cytokines, encompassing interleukin-2 (IL-2), IL-7, IL-15, and IL-21, have been widely used in the development of long-lived adoptively transferred TSCM in vitro. However, challenges, including their non-specific toxicities and off-target effects, have led to substantial efforts for the development of engineered versions to unleash their full potential in the induction and maintenance of T cell stemness in ACT. In this review, we summarize the roles of the four γc family cytokines in the orchestration of adoptively transferred T cell stemness, introduce their engineered versions that modulate TSCM cell formation and demonstrate the potential of their various combinations. Video Abstract.
Collapse
Affiliation(s)
- Mengshi Luo
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjian Gong
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuewen Zhang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huayi Li
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinglei Gao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yong Fang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
43
|
Kang S, Mansurov A, Kurtanich T, Chun HR, Slezak AJ, Volpatti LR, Chang K, Wang T, Alpar AT, Refvik KC, Hansen OI, Borjas GJ, Shim HN, Hultgren KT, Gomes S, Solanki A, Ishihara J, Swartz MA, Hubbell JA. Engineered IL-7 synergizes with IL-12 immunotherapy to prevent T cell exhaustion and promote memory without exacerbating toxicity. SCIENCE ADVANCES 2023; 9:eadh9879. [PMID: 38019919 PMCID: PMC10686557 DOI: 10.1126/sciadv.adh9879] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Cancer immunotherapy is moving toward combination regimens with agents of complementary mechanisms of action to achieve more frequent and robust efficacy. However, compared with single-agent therapies, combination immunotherapies are associated with increased overall toxicity because the very same mechanisms also work in concert to enhance systemic inflammation and promote off-tumor toxicity. Therefore, rational design of combination regimens that achieve improved antitumor control without exacerbated toxicity is a main objective in combination immunotherapy. Here, we show that the combination of engineered, tumor matrix-binding interleukin-7 (IL-7) and IL-12 achieves remarkable anticancer effects by activating complementary pathways without inducing any additive immunotoxicity. Mechanistically, engineered IL-12 provided effector properties to T cells, while IL-7 prevented their exhaustion and boosted memory formation as assessed by tumor rechallenge experiments. The dual combination also rendered checkpoint inhibitor (CPI)-resistant genetically engineered melanoma model responsive to CPI. Thus, our approach provides a framework of evaluation of rationally designed combinations in immuno-oncology and yields a promising therapy.
Collapse
Affiliation(s)
- Seounghun Kang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Aslan Mansurov
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Trevin Kurtanich
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Hye Rin Chun
- Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Anna J. Slezak
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Lisa R. Volpatti
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Kevin Chang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Thomas Wang
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Aaron T. Alpar
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Kirsten C. Refvik
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - O. Isabella Hansen
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Gustavo J. Borjas
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Ha-Na Shim
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Kevin T. Hultgren
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Suzana Gomes
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Ani Solanki
- Animal Resource Center, University of Chicago, Chicago, IL, USA
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, UK
| | - Melody A. Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
- Committee on Immunology, University of Chicago, Chicago, IL, USA
- Committee on Cancer Biology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
44
|
Min H, Valente LA, Xu L, O'Neil SM, Begg LR, Kurtzberg J, Filiano AJ. Improving thymus implantation for congenital athymia with interleukin-7. Clin Transl Immunology 2023; 12:e1475. [PMID: 38020730 PMCID: PMC10665642 DOI: 10.1002/cti2.1475] [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: 03/14/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 12/01/2023] Open
Abstract
Objectives Thymus implantation is a recently FDA-approved therapy for congenital athymia. Patients receiving thymus implantation develop a functional but incomplete T cell compartment. Our objective was to develop a mouse model to study clinical thymus implantation in congenital athymia and to optimise implantation procedures to maximise T cell education and expansion of naïve T cells. Methods Using Foxn1 nu athymic mice as recipients, we tested MHC-matched and -mismatched donor thymi that were implanted as fresh tissue or cultured to remove donor T cells. We first implanted thymus under the kidney capsule and then optimised intramuscular implantation. Using competitive adoptive transfer assays, we investigated whether the failure of newly developed T cells to expand into a complete T cell compartment was because of intrinsic deficits or whether there were deficits in engaging MHC molecules in the periphery. Finally, we tested whether recombinant IL-7 would promote the expansion of host naïve T cells educated by the implanted thymus. Results We determined that thymus implants in Foxn1 nu athymic mice mimic many aspects of clinical thymus implants in patients with congenital athymia. When we implanted cultured, MHC-mismatched donor thymus into Foxn1 nu athymic mice, mice developed a limited T cell compartment with notably underdeveloped naïve populations and overrepresented memory-like T cells. Newly generated T cells were predominantly educated by MHC molecules expressed by the donor thymus, thus potentially undergoing another round of selection once in the peripheral circulation. Using competitive adoptive transfer assays, we compared expansion rates of T cells educated on donor thymus versus T cells educated during typical thymopoiesis in MHC-matched and -mismatched environments. Once in the circulation, regardless of the MHC haplotypes, T cells educated on a donor thymus underwent abnormal expansion with initially more robust proliferation coupled with greater cell death, resembling IL-7 independent spontaneous expansion. Treating implanted mice with recombinant interleukin (IL-7) promoted homeostatic expansion that improved T cell development, expanded the T cell receptor repertoire, and normalised the naïve T cell compartment. Conclusion We conclude that implanting cultured thymus into the muscle of Foxn1 nu athymic mice is an appropriate system to study thymus implantation for congenital athymia and immunodeficiencies. T cells are educated by the donor thymus, yet naïve T cells have deficits in expansion. IL-7 greatly improves T cell development after thymus implantation and may offer a novel strategy to improve outcomes of clinical thymus implantation.
Collapse
Affiliation(s)
- Hyunjung Min
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Laura A Valente
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
- Department of PathologyDuke UniversityDurhamNCUSA
| | - Li Xu
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Shane M O'Neil
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Lauren R Begg
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Joanne Kurtzberg
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
- Department of PediatricsDuke UniversityDurhamNCUSA
| | - Anthony J Filiano
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
- Department of PathologyDuke UniversityDurhamNCUSA
- Department of NeurosurgeryDuke UniversityDurhamNCUSA
- Department of ImmunologyDuke UniversityDurhamNCUSA
| |
Collapse
|
45
|
Salimi A, Schemionek‐Reinders M, Huber M, Vieri M, Patterson JB, Alten J, Brümmendorf TH, Kharabi Masouleh B, Appelmann I. XBP1 promotes NRAS G12D pre-B acute lymphoblastic leukaemia through IL-7 receptor signalling and provides a therapeutic vulnerability for oncogenic RAS. J Cell Mol Med 2023; 27:3363-3377. [PMID: 37753803 PMCID: PMC10623536 DOI: 10.1111/jcmm.17904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 09/28/2023] Open
Abstract
Activating point mutations of the RAS gene act as driver mutations for a subset of precursor-B cell acute lymphoblastic leukaemias (pre-B ALL) and represent an ambitious target for therapeutic approaches. The X box-binding protein 1 (XBP1), a key regulator of the unfolded protein response (UPR), is critical for pre-B ALL cell survival, and high expression of XBP1 confers poor prognosis in ALL patients. However, the mechanism of XBP1 activation has not yet been elucidated in RAS mutated pre-B ALL. Here, we demonstrate that XBP1 acts as a downstream linchpin of the IL-7 receptor signalling pathway and that pharmacological inhibition or genetic ablation of XBP1 selectively abrogates IL-7 receptor signalling via inhibition of its downstream effectors, JAK1 and STAT5. We show that XBP1 supports malignant cell growth of pre-B NRASG12D ALL cells and that genetic loss of XBP1 consequently leads to cell cycle arrest and apoptosis. Our findings reveal that active XBP1 prevents the cytotoxic effects of a dual PI3K/mTOR pathway inhibitor (BEZ235) in pre-B NRASG12D ALL cells. This implies targeting XBP1 in combination with BEZ235 as a promising new targeted strategy against the oncogenic RAS in NRASG12D -mutated pre-B ALL.
Collapse
Affiliation(s)
- Azam Salimi
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging CenterEberhard Karls University TübingenTübingenGermany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies"TübingenGermany
| | - Mirle Schemionek‐Reinders
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Michael Huber
- Medical Faculty, Institute of Biochemistry and Molecular ImmunologyRWTH Aachen UniversityAachenGermany
| | - Margherita Vieri
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
| | | | - Julia Alten
- Department of PediatricsUniversity Medical Centre Schleswig‐HolsteinKielGermany
| | - Tim H. Brümmendorf
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Behzad Kharabi Masouleh
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
| | - Iris Appelmann
- Department of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation, Medical FacultyRWTH Aachen UniversityAachenGermany
| |
Collapse
|
46
|
Dower WJ, Park AI, Bakker AV, Cwirla SE, Pongtornpipat P, Williams BM, Joshi P, Baxter BA, Needels MC, Barrett RW. A mechanistically novel peptide agonist of the IL-7 receptor that addresses limitations of IL-7 cytokine therapy. PLoS One 2023; 18:e0286834. [PMID: 37874823 PMCID: PMC10597491 DOI: 10.1371/journal.pone.0286834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/06/2023] [Indexed: 10/26/2023] Open
Abstract
Interleukin (IL)-7 is broadly active on T-cell populations, and modified versions have been clinically evaluated for a variety of therapeutic applications, including cancer, lymphopenia, and infectious diseases; and found to be relatively well-tolerated and biologically active. Here we describe novel IL-7R agonists that are unrelated in structure to IL-7, bind to the receptor subunits differently from IL-7, but closely emulate IL-7 biology. The small size, low structural complexity, and the natural amino acid composition of the pharmacologically active peptide MDK1472 allows facile incorporation into protein structures, such as the IgG2-Fc fusion MDK-703. This molecule possesses properties potentially better suited to therapeutic applications than native IL-7 or its derivatives. We compared these compounds with IL-7 for immune cell selectivity, induction of IL-7R signaling, receptor-mediated internalization, proliferation, and generation of immune cell phenotypes in human and non-human primate (NHP) peripheral blood cells in vitro; and found them to be similar in biological activity to IL-7. In cynomolgus macaques, MDK-703 exhibits a circulating half-life of 46 hr and produces sustained T-cell expansion characteristic of IL-7 treatment. In the huCD34+-engrafted NSG mouse model of the human immune system, MDK-703 induces an immune cell profile very similar to that generated by IL-7-derived compounds; including the pronounced expansion of memory T-cells, particularly the population of stem-like memory T-cells (Tscm) which may be important for anti-tumor activities reported with IL-7 treatment. Clinical administration of IL-7 and modified variants has been reported to induce anti-drug antibodies (ADAs), including IL-7 neutralizing antibodies. The novel peptide agonist reported here scores very low in predicted immunogenicity, and because the peptide lacks sequence similarity with IL-7, the problematic immunogenic neutralization of endogenous cytokine should not occur. The properties we report here implicate MDK-703 as a candidate for clinical evaluation in oncology, anti-viral and other infectious disease, vaccine enhancement, and treatment of lymphopenia.
Collapse
Affiliation(s)
- William J. Dower
- Medikine, Inc., Menlo Park, California, United States of America
| | | | - Alice V. Bakker
- Medikine, Inc., Menlo Park, California, United States of America
| | - Steven E. Cwirla
- Medikine, Inc., Menlo Park, California, United States of America
| | | | - Blake M. Williams
- Medikine, Inc., Menlo Park, California, United States of America
- Department of Biomedical Engineering, and Center for Quantitative Bioinformatics and Quantitative Biology, Colleges of Engineering and Medicine, University of Illinois Chicago, IL, United States of America
| | - Prarthana Joshi
- Medikine, Inc., Menlo Park, California, United States of America
| | - Bryan A. Baxter
- Medikine, Inc., Menlo Park, California, United States of America
| | | | | |
Collapse
|
47
|
Tripathi S, Tsang JS, Park K. Systems immunology of regulatory T cells: can one circuit explain it all? Trends Immunol 2023; 44:766-781. [PMID: 37690962 PMCID: PMC10543564 DOI: 10.1016/j.it.2023.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 09/12/2023]
Abstract
Regulatory T (Treg) cells play vital roles in immune homeostasis and response, including discrimination between self- and non-self-antigens, containment of immunopathology, and inflammation resolution. These diverse functions are orchestrated by cellular circuits involving Tregs and other cell types across space and time. Despite dramatic progress in our understanding of Treg biology, a quantitative framework capturing how Treg-containing circuits give rise to these diverse functions is lacking. Here, we propose that different facets of Treg function can be interpreted as distinct operating regimes of the same underlying circuit. We discuss how a systems immunology approach, involving quantitative experiments, computational modeling, and machine learning, can advance our understanding of Treg function, and help identify general operating and design principles underlying immune regulation.
Collapse
Affiliation(s)
- Shubham Tripathi
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA.
| | - John S Tsang
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA; Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA.
| | - Kyemyung Park
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA; Graduate School of Health Science and Technology and Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea.
| |
Collapse
|
48
|
Rahman T, Das A, Abir MH, Nafiz IH, Mahmud AR, Sarker MR, Emran TB, Hassan MM. Cytokines and their role as immunotherapeutics and vaccine Adjuvants: The emerging concepts. Cytokine 2023; 169:156268. [PMID: 37320965 DOI: 10.1016/j.cyto.2023.156268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023]
Abstract
Cytokines are a protein family comprising interleukins, lymphokines, chemokines, monokines and interferons. They are significant constituents of the immune system, and they act in accordance with specific cytokine inhibiting compounds and receptors for the regulation of immune responses. Cytokine studies have resulted in the establishment of newer therapies which are being utilized for the treatment of several malignant diseases. The advancement of these therapies has occurred from two distinct strategies. The first strategy involves administrating the recombinant and purified cytokines, and the second strategy involves administrating the therapeutics which inhibits harmful effects of endogenous and overexpressed cytokines. Colony stimulating factors and interferons are two exemplary therapeutics of cytokines. An important effect of cytokine receptor antagonist is that they can serve as anti-inflammatory agents by altering the treatments of inflammation disorder, therefore inhibiting the effects of tumour necrosis factor. In this article, we have highlighted the research behind the establishment of cytokines as therapeutics and vaccine adjuvants, their role of immunotolerance, and their limitations.
Collapse
Affiliation(s)
- Tanjilur Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Ayan Das
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Mehedy Hasan Abir
- Faculty of Food Science and Technology, Chattogram Veterinary and Animal Sciences University, Chattogram 4225, Bangladesh
| | - Iqbal Hossain Nafiz
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chattogram 4331, Bangladesh
| | - Aar Rafi Mahmud
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Rifat Sarker
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chattogram 4381, Bangladesh; Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Mohammad Mahmudul Hassan
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Chattogram 4225, Bangladesh; Queensland Alliance for One Health Sciences, School of Veterinary Science, The University of Queensland, Queensland 4343, Australia.
| |
Collapse
|
49
|
Leśnik P, Janc J, Mierzchala-Pasierb M, Tański W, Wierciński J, Łysenko L. Interleukin-7 and interleukin-15 as prognostic biomarkers in sepsis and septic shock: Correlation with inflammatory markers and mortality. Cytokine 2023; 169:156277. [PMID: 37348189 DOI: 10.1016/j.cyto.2023.156277] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023]
Abstract
Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection and a syndrome shaped by pathogen and host factors evolving over time. During sepsis, the absolute number of lymphocytes decreases. CD4+ and CD8+ T cells, B cells, and NK cells are reduced. Lymphocytes are an essential element of the body's defence against pathogens. Interleukin 7 has strong anti-apoptotic properties and induces the proliferation of CD4+ and CD8+ T lymphocytes. IL-15 prompts the generation of mature NK cells in the bone marrow, plays an important role in the generation, cytotoxicity, and survival of CD8+ T lymphocytes, and is essential for the survival of natural killer T (NKT) and intestinal intraepithelial lymphocytes (IELs). The study highlights the importance of monitoring IL-7 levels in patients with sepsis and septic shock, as low levels of this cytokine were associated with an increased risk of mortality. Physicians should consider using IL-7 levels as a biomarker to identify patients who are at higher risk of mortality and may require more aggressive treatment.
Collapse
Affiliation(s)
- Patrycja Leśnik
- Department of Anaesthesiology and Intensive Therapy, 4th Military Clinical Hospital, 50-981 Wroclaw, Poland.
| | - Jarosław Janc
- Department of Anaesthesiology and Intensive Therapy, 4th Military Clinical Hospital, 50-981 Wroclaw, Poland.
| | | | - Wojciech Tański
- Department of Internal Medicine, 4th Military Clinical Hospital, 50-981 Wroclaw, Poland.
| | - Jan Wierciński
- Department of Anaesthesiology and Intensive Therapy, 4th Military Clinical Hospital, 50-981 Wroclaw, Poland.
| | - Lidia Łysenko
- Department of Anaesthesiology and Intensive Therapy, Wroclaw Medical University, 50-556 Wroclaw, Poland.
| |
Collapse
|
50
|
Gallardo-Dodd CJ, Oertlin C, Record J, Galvani RG, Sommerauer C, Kuznetsov NV, Doukoumopoulos E, Ali L, Oliveira MMS, Seitz C, Percipalle M, Nikić T, Sadova AA, Shulgina SM, Shmarov VA, Kutko OV, Vlasova DD, Orlova KD, Rykova MP, Andersson J, Percipalle P, Kutter C, Ponomarev SA, Westerberg LS. Exposure of volunteers to microgravity by dry immersion bed over 21 days results in gene expression changes and adaptation of T cells. SCIENCE ADVANCES 2023; 9:eadg1610. [PMID: 37624890 PMCID: PMC10456848 DOI: 10.1126/sciadv.adg1610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 07/25/2023] [Indexed: 08/27/2023]
Abstract
The next steps of deep space exploration are manned missions to Moon and Mars. For safe space missions for crew members, it is important to understand the impact of space flight on the immune system. We studied the effects of 21 days dry immersion (DI) exposure on the transcriptomes of T cells isolated from blood samples of eight healthy volunteers. Samples were collected 7 days before DI, at day 7, 14, and 21 during DI, and 7 days after DI. RNA sequencing of CD3+ T cells revealed transcriptional alterations across all time points, with most changes occurring 14 days after DI exposure. At day 21, T cells showed evidence of adaptation with a transcriptional profile resembling that of 7 days before DI. At 7 days after DI, T cells again changed their transcriptional profile. These data suggest that T cells adapt by rewiring their transcriptomes in response to simulated weightlessness and that remodeling cues persist when reexposed to normal gravity.
Collapse
Affiliation(s)
- Carlos J. Gallardo-Dodd
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Christian Oertlin
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Julien Record
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Rômulo G. Galvani
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Laboratory of Bioinformatics and Computational Biology, Division of Experimental and Translational Research, Brazilian National Cancer Institute (INCA), Rio de Janeiro, RJ, Brazil
- Universidade Veiga de Almeida, Rio de Janeiro, Brazil
- Laboratory for Thymus Research (LPT), Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil
| | - Christian Sommerauer
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Nikolai V. Kuznetsov
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Russian Federation State Research Center Institute of Biomedical Problems RAS, Moscow, Russia
| | | | - Liaqat Ali
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
- Core Technology Platform, NYUAD, Abu Dhabi, United Arab Emirates
| | - Mariana M. S. Oliveira
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christina Seitz
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mathias Percipalle
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Tijana Nikić
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia A. Sadova
- Russian Federation State Research Center Institute of Biomedical Problems RAS, Moscow, Russia
| | - Sofia M. Shulgina
- Russian Federation State Research Center Institute of Biomedical Problems RAS, Moscow, Russia
| | - Vjacheslav A. Shmarov
- Russian Federation State Research Center Institute of Biomedical Problems RAS, Moscow, Russia
| | - Olga V. Kutko
- Russian Federation State Research Center Institute of Biomedical Problems RAS, Moscow, Russia
| | - Daria D. Vlasova
- Russian Federation State Research Center Institute of Biomedical Problems RAS, Moscow, Russia
| | - Kseniya D. Orlova
- Russian Federation State Research Center Institute of Biomedical Problems RAS, Moscow, Russia
| | - Marina P. Rykova
- Russian Federation State Research Center Institute of Biomedical Problems RAS, Moscow, Russia
| | - John Andersson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Piergiorgio Percipalle
- Program in Biology, Division of Science and Mathematics, New York University Abu Dhabi (NYUAD), Abu Dhabi, United Arab Emirates
- Center for Genomics and Systems Biology, NYUAD, Abu Dhabi, United Arab Emirates
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Claudia Kutter
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Sergey A. Ponomarev
- Russian Federation State Research Center Institute of Biomedical Problems RAS, Moscow, Russia
| | - Lisa S. Westerberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|