1
|
Kim GD, Shin SI, Sun P, Lee JE, Chung C, Kang YE, Kang DH, Park J. Single-cell RNA sequencing of baseline PBMCs predicts ICI efficacy and irAE severity in patients with NSCLC. J Immunother Cancer 2025; 13:e011636. [PMID: 40404203 DOI: 10.1136/jitc-2025-011636] [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: 04/29/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have transformed treatment and have provided significant clinical benefits and durable responses for patients with advanced non-small cell lung cancer (NSCLC). However, only a small percentage of patients respond to ICI treatment, and immune-related adverse events (irAEs) leading to treatment discontinuation remain challenging. Despite the recognized need for biomarkers to predict both the efficacy of ICIs and the risk of irAEs, such biomarkers are yet to be clearly identified. METHODS In this study, we performed single-cell RNA sequencing (scRNA-seq) of peripheral blood mononuclear cells (PBMCs) from 33 patients with NSCLC before ICIs treatment. To validate our findings, we reanalyzed public scRNA-seq data, conducted a cytometric bead array (CBA), and supported our findings with T-cell receptor sequencing. RESULTS While the immune response was more pronounced in patients with a favorable prognosis, the hypoxic pathway was more prominent in patients with primary resistance. Lymphocytes such as CD8 T cells, CD4 T cells, and natural killer cells were primarily involved in these pathways, with PRF1 and GZMB expression showing strong associations with favorable prognosis. In contrast, irAEs were mainly linked to myeloid cells, such as monocytes and macrophages. As irAE severity increased, inflammation and the TNF-NFKB1 pathway were more prominent. Specifically, increased expression of IL1B, CXCL8, and CXCL2 in monocytes and TNF in macrophages was closely associated with severe irAE through involvement in these pathways.Notably, the increase of PRF1 and GZMB expression showed a close association with both a favorable prognosis and a reduced severity of irAE, which was validated through CBA analysis. Moreover, the expression of these key markers varied according to prognosis and irAE severity regardless of patient background, such as programmed death-ligand 1 expression levels, tumor histology, or prior treatment regimens. CONCLUSIONS This study identified biological pathways and key biomarkers associated with ICI prognosis and irAE severity using PBMC samples before treatment. These findings provide a foundation for improved therapeutic strategies that enhance clinical outcomes while minimizing ICI treatment-associated risks.
Collapse
Affiliation(s)
- Gyeong Dae Kim
- Life Science, Gwangju Institute of Science and Technology, Gwangju, Buk-gu, Korea (the Republic of)
| | - So-I Shin
- Life Science, Gwangju Institute of Science and Technology, Gwangju, Buk-gu, Korea (the Republic of)
| | - Pureum Sun
- Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jeong Eun Lee
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Chaeuk Chung
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Yea Eun Kang
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Da Hyun Kang
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jihwan Park
- Life Science, Gwangju Institute of Science and Technology, Gwangju, Buk-gu, Korea (the Republic of)
| |
Collapse
|
2
|
Hammerstad SS, Lee HJ, Tomer Y, Stefan-Lifshitz M. Immune checkpoint inhibitors associated thyroiditis: mechanisms and clinical outcomes. J Endocrinol Invest 2025:10.1007/s40618-025-02608-2. [PMID: 40353948 DOI: 10.1007/s40618-025-02608-2] [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: 12/01/2024] [Accepted: 05/06/2025] [Indexed: 05/14/2025]
Abstract
Background Immune checkpoint inhibitors (ICI) have revolutionized cancer treatment improving prognosis in many cancers. However, ICI often trigger immune-related adverse events (irAEs) that can affect various organ systems, leading to significant clinical challenges. The most frequent irAEs are those affecting the endocrine glands, reported in approximately 10% of treated patients. Thyroid dysfunction is the most common endocrine irAEs, mainly associated with PD-1/PD-L1 blockade therapies. Transient asymptomatic thyrotoxicosis is the most common form of clinical presentation, often followed by hypothyroidism. Interestingly, several studies have demonstrated that irAEs correlate with the response to cancer therapy and with improved overall survival (OS). The mechanisms underlying thyroid irAEs are not fully elucidated but complex interactions between genetic predisposition to thyroid autoimmunity, distinct immune mechanisms and thyroid cell intrinsic mechanisms are thought to drive thyroiditis associated with ICI therapy. Purpose In this review we discuss the latest data on clinical features of thyroid irAEs, proposed mechanisms and their association with improved survival.
Collapse
Affiliation(s)
- Sara S Hammerstad
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Oslo, 4950, 0424, PB, Norway
- Institute of Clinical Medicine, University of Oslo, Norway Klaus Torgårdsvei 3, Oslo, 0372, Norway
| | - Hanna J Lee
- Department of Medicine, The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Yaron Tomer
- Department of Medicine, The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Mihaela Stefan-Lifshitz
- Department of Medicine, The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| |
Collapse
|
3
|
Paiola M, Portnoy DM, Hao LY, Bukhari S, Winchester RJ, Henick BS, Mor A, Gartshteyn Y. Osteoarthritis increases the risk of inflammatory arthritis due to immune checkpoint inhibitors associated with tissue-resident memory T cells. J Immunother Cancer 2025; 13:e010758. [PMID: 40118498 PMCID: PMC11931944 DOI: 10.1136/jitc-2024-010758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/18/2025] [Indexed: 03/23/2025] Open
Abstract
OBJECTIVE Immune checkpoint inhibitors (ICIs) have significantly advanced cancer treatment, but they can also lead to immune-related adverse events (irAEs), including inflammatory arthritis. Understanding the risk factors and underlying mechanisms of irAE pathogenesis is crucial for optimal patient management. Increasing evidence suggests that ICI-mediated activation of tissue-resident memory T cells (TRM) significantly eliminates cancer cells and is associated with irAE-related colitis and dermatitis. However, it remains unknown why the development of these irAEs is restricted to a subset of patients. We hypothesized that osteoarthritis (OA) associated tissue damage and chronic inflammation lead to the recruitment and differentiation of joint TRM cells, predisposing individuals to ICI-induced arthritis. METHODS Using a comprehensive approach, we compared the prevalence of OA in patients with irAE-arthritis to those with irAE non-arthritis and those without irAEs. Additionally, we used advanced immunophenotyping techniques to characterize T-cell populations in the blood and synovial fluid of patients with OA and irAE-arthritis. RESULTS Our findings revealed a significantly higher prevalence of OA in patients who developed irAE-arthritis than controls. Furthermore, the multivariable analysis identified OA, body mass index, and smoking as independent risk factors for the development of irAE-arthritis. TRM cells expressing programmed cell death protein-1 (PD-1) were the predominant synovial T cells in OA joints. These cells were directly targeted by ICIs, resulting in an inflammatory immune response and the transition from OA to irAE-arthritis. CONCLUSION This study, the first of its kind, identifies OA as a significant risk factor for irAEarthritis. It reveals a potential mechanism by which ICIs activate PD-1-positive TRM cells in OA joints, resulting in tissue inflammation and irAE-arthritis. This research could significantly enhance the management and treatment of patients with cancer receiving ICIs.
Collapse
Affiliation(s)
- Matthieu Paiola
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
| | - Daniel M Portnoy
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
- Department of Medicine, Division of Rheumatology, Columbia University Medical Center, New York, New York, USA
| | - Luke Yi Hao
- Columbia University Vagelos College of Physicians and Surgeons, New York, New York, USA
| | - Shoiab Bukhari
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
| | - Robert J Winchester
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
- Department of Medicine, Division of Rheumatology, Columbia University Irving Medical Center, New York, New York, USA
| | - Brian S Henick
- Herbert Irvine Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Adam Mor
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
- Herbert Irvine Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Yevgeniya Gartshteyn
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, USA
- Department of Medicine, Division of Rheumatology, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
4
|
Wang S, Xu D, Xiao L, Liu B, Yuan X. Radiation-induced lung injury: from mechanism to prognosis and drug therapy. Radiat Oncol 2025; 20:39. [PMID: 40082925 PMCID: PMC11907960 DOI: 10.1186/s13014-025-02617-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
Radiation induced lung injury, known as the main complication of thoracic radiation, remains to be a major resistance to tumor treatment. Based on the recent studies on radiation-induced lung injury, this review collated the possible mechanisms at the level of target cells and key pathways, corresponding prognostic models including predictors, patient size, number of centers, radiotherapy technology, construction methods and accuracy, and pharmacotherapy including drugs, targets, therapeutic effects, impact on anti-tumor treatment and research types. The research priorities and limitations are summarized to provide a reference for the research and management of radiation-induced lung injury.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, Jiangsu, 210000, China
| | - Duo Xu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Lingyan Xiao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
5
|
Martini E, Cremonesi M, Felicetta A, Serio S, Puccio S, Pelamatti E, van Beek JJP, Papadopoulou V, Catalano C, Fanuele F, Giuliano D, Basso G, Bonfiglio CA, Panico C, Vacchiano M, Carullo P, Papa L, D'Andrea C, Tuzger N, Marchini S, Magistroni P, Deaglio S, Amoroso A, Lugli E, Condorelli G, Kallikourdis M. Autoimmune-Like Mechanism in Heart Failure Enables Preventive Vaccine Therapy. Circ Res 2025; 136:4-25. [PMID: 39629560 DOI: 10.1161/circresaha.124.324999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/31/2024] [Accepted: 11/20/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Heart failure (HF) is strongly associated with inflammation. In pressure overload (PO)-induced HF, cardiac stress triggers adaptive immunity, ablation or inhibition of which blocks disease progression. We hypothesized that PO-HF might fulfill the often-used criteria of autoimmunity: if so, the associated adaptive immune response would be not only necessary but also sufficient to induce HF; it should also be possible to identify self-antigens driving the autoimmune response. Finally, we hypothesized that such an antigen-specific response can be manipulated to preventively reduce the severity of PO-HF in a tolerizing vaccine. METHODS We used the transfer of lymphocytes or serum from PO-HF mice into healthy recipients to assess whether the adaptive response is sufficient to induce disease. We devised a novel pipeline to identify self-antigens driving the response. We immunized healthy mice with novel antigens to assess whether they induce disease. To determine whether these antigens could be present in human patients, we sought to detect existing responses against these antigens in patients with HF. Finally, we used the antigens in an oral tolerance protocol to preventively protect mice from subsequently induced PO-HF, analyzing the results with next-generation sequencing. RESULTS We found that PO-HF fulfills the criteria of an autoimmune disease, albeit partially, and identified novel cardiac self-antigens, capable of inducing cardiac dysfunction. The novel antigens in a tolerizing vaccine formulation preemptively reduced the severity of disease triggered by subsequent application of PO, via induction of effector regulatory T cells, enabling a potent reduction of PO-driven loss of systolic function, cardiac inflammation, and proinflammatory CD4+ T-cell clonal expansion. CONCLUSIONS We demonstrate that PO-HF is triggered by hemodynamic stress and then sets off an autoimmune-like response against cardiac self-antigens. The antigens can be used to reduce the severity of future-onset disease, via oral tolerization, effectively acting as a protective vaccine.
Collapse
Affiliation(s)
- Elisa Martini
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
| | - Marco Cremonesi
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
| | - Arianna Felicetta
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Simone Serio
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Institute of Genetics and Biomedical Research, Milan Unit, Consiglio Nazionale delle Ricerche, Italy (S.S., S.P.)
| | - Simone Puccio
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy (S.P., J.J.P.v.B., E.L.)
- Institute of Genetics and Biomedical Research, Milan Unit, Consiglio Nazionale delle Ricerche, Italy (S.S., S.P.)
| | - Erica Pelamatti
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
| | - Jasper J P van Beek
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy (S.P., J.J.P.v.B., E.L.)
| | - Vasiliki Papadopoulou
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Chiara Catalano
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Francesca Fanuele
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Desirée Giuliano
- Humanitas Genomics Facility, IRCCS Humanitas Research Hospital, Rozzano, Italy (D.G., G.B., S.M.)
| | - Gianluca Basso
- Humanitas Genomics Facility, IRCCS Humanitas Research Hospital, Rozzano, Italy (D.G., G.B., S.M.)
| | - Cecilia Assunta Bonfiglio
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität München, Germany (C.A.B.)
| | - Cristina Panico
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Marco Vacchiano
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Pierluigi Carullo
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Laura Papa
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
| | - Carla D'Andrea
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Naz Tuzger
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Sergio Marchini
- Humanitas Genomics Facility, IRCCS Humanitas Research Hospital, Rozzano, Italy (D.G., G.B., S.M.)
| | - Paola Magistroni
- Immunogenetics and Transplant Biology Service, Città della Salute e della Scienza Hospital, Turin, Italy (P.M., S.D., A.A.)
| | - Silvia Deaglio
- Immunogenetics and Transplant Biology Service, Città della Salute e della Scienza Hospital, Turin, Italy (P.M., S.D., A.A.)
- Department of Medical Sciences, University of Turin, Italy (S.D., A.A.)
| | - Antonio Amoroso
- Immunogenetics and Transplant Biology Service, Città della Salute e della Scienza Hospital, Turin, Italy (P.M., S.D., A.A.)
- Department of Medical Sciences, University of Turin, Italy (S.D., A.A.)
| | - Enrico Lugli
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy (S.P., J.J.P.v.B., E.L.)
| | - Gianluigi Condorelli
- Cardio Center, IRCCS Humanitas Research Hospital, Rozzano, Italy (A.F., S.S., C.P., M.V., P.C., L.P., C.D., G.C.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| | - Marinos Kallikourdis
- Adaptive Immunity Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Rozzano, Italy (E.M., M.C., E.P., V.P., C.C., F.F., C.A.B., N.T., M.K.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy (V.P., C.C., F.F., C.P., C.D., N.T., G.C., M.K.)
| |
Collapse
|
6
|
Xiong J, Chen G, Lin B, Zhong L, Jiang X, Lu H. Integrative analysis of single-Cell RNA sequencing and experimental validation in the study of abdominal aortic aneurysm progression. Gene 2024; 929:148820. [PMID: 39103059 DOI: 10.1016/j.gene.2024.148820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/13/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Abdominal aortic aneurysm (AAA) is a complex vascular disorder characterized by the progressive dilation of the abdominal aorta, with a high risk of rupture and mortality. Understanding the cellular interactions and molecular mechanisms underlying AAA development is critical for identifying potential therapeutic targets. METHODS This study utilized datasets GSE197748, GSE164678 and GSE183464 from the GEO database, encompassing bulk and single-cell RNA sequencing data from AAA and control samples. We performed principal component analysis, differential expression analysis, and functional enrichment analysis to identify key pathways involved in AAA. Cell-cell interactions were investigated using CellPhoneDB, focusing on fibroblasts, vascular smooth muscle cells (VSMCs), and macrophages. We further validated our findings using a mouse model of AAA induced by porcine pancreatic enzyme infusion, followed by gene expression analysis and co-immunoprecipitation experiments. RESULTS Our analysis revealed significant alterations in gene expression profiles between AAA and control samples, with a pronounced immune response and cell adhesion pathways being implicated. Single-cell RNA sequencing data highlighted an increased proportion of pro-inflammatory macrophages, along with changes in the composition of fibroblasts and VSMCs in AAA. CellPhoneDB analysis identified critical ligand-receptor interactions, notably collagen type I alpha 1 chain (COL1A1)/COL1A2-CD18 and thrombospondin 1 (THBS1)-CD3, suggesting complex communication networks between fibroblasts and VSMCs. In vivo experiments confirmed the upregulation of these genes in AAA mice and demonstrated the functional interaction between COL1A1/COL1A2 and CD18. CONCLUSION The interaction between fibroblasts and VSMCs, mediated by specific ligand-receptor pairs such as COL1A1/COL1A2-CD18 and THBS1-CD3, plays a pivotal role in AAA pathogenesis.
Collapse
MESH Headings
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Animals
- Mice
- Single-Cell Analysis/methods
- Humans
- Sequence Analysis, RNA/methods
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Macrophages/metabolism
- Disease Progression
- Fibroblasts/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Male
- Mice, Inbred C57BL
- Gene Expression Profiling/methods
- Cell Communication/genetics
- Collagen Type I/genetics
- Collagen Type I/metabolism
Collapse
Affiliation(s)
- Jie Xiong
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China
| | - Guojun Chen
- Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou 510515, China
| | - Beiyou Lin
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China
| | - Lintao Zhong
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China
| | - Xiaofei Jiang
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China.
| | - Hongyun Lu
- Department of Cardiology, Zhuhai Hospital affiliated with Jinan University (Zhuhai People's Hospital), Zhuhai 519000, China.
| |
Collapse
|
7
|
Huang N, Ortega J, Kimbrell K, Lee J, Scott LN, Peluso EM, Wang SJ, Kao E, Kim K, Olay J, Quandt Z, Angell TE, Su MA, Lechner MG. Polyfunctional IL-21 + IFNG + T follicular helper cells contribute to checkpoint inhibitor diabetes mellitus and can be targeted by JAK inhibitor therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625710. [PMID: 39677814 PMCID: PMC11642801 DOI: 10.1101/2024.11.27.625710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Immune checkpoint inhibitors (ICI) have revolutionized cancer therapy, but their use is limited by the development of autoimmunity in healthy tissues as a side effect of treatment. Such immune-related adverse events (IrAE) contribute to hospitalizations, cancer treatment interruption and even premature death. ICI-induced autoimmune diabetes mellitus (ICI-T1DM) is a life-threatening IrAE that presents with rapid pancreatic beta-islet cell destruction leading to hyperglycemia and life-long insulin dependence. While prior reports have focused on CD8+ T cells, the role for CD4+ T cells in ICI-T1DM is less understood. Here, we identify expansion CD4+ T follicular helper (Tfh) cells expressing interleukin 21 (IL-21) and interferon gamma (IFNG) as a hallmark of ICI-T1DM. Furthermore, we show that both IL-21 and IFNG are critical cytokines for autoimmune attack in ICI-T1DM. Because IL-21 and IFNG both signal through JAK-STAT pathways, we reasoned that JAK inhibitors (JAKi) may protect against ICI-T1DM. Indeed, JAKi provide robust in vivo protection against ICI-T1DM in a mouse model that is associated with decreased islet-infiltrating Tfh cells. Moreover, JAKi therapy impaired Tfh cell differentiation in patients with ICI-T1DM. These studies highlight CD4+ Tfh cells as underrecognized but critical mediators of ICI-T1DM that may be targeted with JAKi to prevent this grave IrAE.
Collapse
Affiliation(s)
- Nicole Huang
- Division of Endocrinology, Diabetes, and Metabolism, University of California Los Angeles (UCLA) David Geffen School of Medicine, Los Angeles, CA 90095
| | | | - Kyleigh Kimbrell
- Division of Endocrinology, Diabetes, and Metabolism, University of California Los Angeles (UCLA) David Geffen School of Medicine, Los Angeles, CA 90095
| | - Joah Lee
- Division of Endocrinology, Diabetes, and Metabolism, University of California Los Angeles (UCLA) David Geffen School of Medicine, Los Angeles, CA 90095
| | | | - Esther M. Peluso
- UCLA/California Institute of Technology Medical Scientist Training Program, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
| | - Sarah J. Wang
- Division of Endocrinology, Diabetes, and Metabolism, University of California Los Angeles (UCLA) David Geffen School of Medicine, Los Angeles, CA 90095
| | - Ellie Kao
- California State Polytechnic University, Pomona, CA 91768
| | - Kristy Kim
- Division of Endocrinology, Diabetes, and Metabolism, University of California Los Angeles (UCLA) David Geffen School of Medicine, Los Angeles, CA 90095
| | - Jarod Olay
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
| | - Zoe Quandt
- Division of Endocrinology and Metabolism, University of California San Francisco Medical School, San Francisco, CA 94143
| | - Trevor E. Angell
- Division of Endocrinology and Diabetes, University of Southern California Keck School of Medicine; Los Angeles, CA 90033
| | - Maureen A. Su
- Department of Microbiology, Immunology, and Molecular Genetics, UCLA David Geffen School of Medicine, Los Angeles, CA 90095
- Division of Pediatric Endocrinology, UCLA David Geffen School of Medicine; Los Angeles, CA 90095
| | - Melissa G. Lechner
- Division of Endocrinology, Diabetes, and Metabolism, University of California Los Angeles (UCLA) David Geffen School of Medicine, Los Angeles, CA 90095
| |
Collapse
|
8
|
Chang CJ, Hsu CY, Liu Q, Shyr Y. VICTOR: Validation and inspection of cell type annotation through optimal regression. Comput Struct Biotechnol J 2024; 23:3270-3280. [PMID: 39296808 PMCID: PMC11408377 DOI: 10.1016/j.csbj.2024.08.028] [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: 05/08/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/21/2024] Open
Abstract
Single-cell RNA sequencing provides unprecedent opportunities to explore the heterogeneity and dynamics inherent in cellular biology. An essential step in the data analysis involves the automatic annotation of cells. Despite development of numerous tools for automated cell annotation, assessing the reliability of predicted annotations remains challenging, particularly for rare and unknown cell types. Here, we introduce VICTOR: Validation and inspection of cell type annotation through optimal regression. VICTOR aims to gauge the confidence of cell annotations by an elastic-net regularized regression with optimal thresholds. We demonstrated that VICTOR performed well in identifying inaccurate annotations, surpassing existing methods in diagnostic ability across various single-cell datasets, including within-platform, cross-platform, cross-studies, and cross-omics settings.
Collapse
Affiliation(s)
- Chia-Jung Chang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN 37203, USA
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Yuan Hsu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Qi Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203, USA
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| |
Collapse
|
9
|
Verheye E, Kancheva D, Satilmis H, Vandewalle N, Fan R, Bardet PMR, Clappaert EJ, Verstaen K, De Becker A, Vanderkerken K, De Veirman K, Laoui D. A single-cell transcriptomic map of the murine and human multiple myeloma immune microenvironment across disease stages. J Hematol Oncol 2024; 17:107. [PMID: 39511632 PMCID: PMC11546219 DOI: 10.1186/s13045-024-01629-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/26/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND The long-term effectiveness of immunotherapies against Multiple Myeloma (MM) remains elusive, demonstrated by the inevitable relapse in patients. This underscores the urgent need for an in-depth analysis of the MM tumor-immune microenvironment (TME). Hereto, a representative immunocompetent MM mouse model can offer a valuable approach to study the dynamic changes within the MM-TME and to uncover potential resistance mechanisms hampering effective and durable therapeutic strategies in MM. METHODS We generated a comprehensive single-cell RNA-sequencing atlas of the MM-TME in bone marrow and spleen encompassing different stages of disease, using the immunocompetent 5T33MM mouse model. Through comparative analysis, we correlated our murine dataset with the pathogenesis in MM patients by reanalyzing publicly available datasets of human bone marrow samples across various disease stages. Using flow cytometry, we validated the dynamic changes upon disease progression in the 5T33MM model. Furthermore, interesting target populations, as well as the immune-boosting anti-CD40 agonist (αCD40) therapy were tested ex vivo on murine and human primary samples and in vivo using the 5T33MM model. RESULTS In this study, we identified the heterogenous and dynamic changes within the TME of murine and human MM. We found that the MM-TME was characterized by an increase in T cells, accompanied with an exhausted phenotype. Although neutrophils appeared to be rather innocuous at early disease stages, they acquired a pro-tumorigenic phenotype during MM progression. Moreover, conventional dendritic cells (cDCs) showed a less activated phenotype in MM, underscoring the potential of immune-boosting therapies such as αCD40 therapy. Importantly, we provided the first pre-clinical evaluation of αCD40 therapy and demonstrated successful induction of cDC- and T-cell activation, accompanied by a significant short-term anti-tumor response. CONCLUSIONS This resource provides a comprehensive and detailed immune atlas of the evolution in human and murine MM disease progression. Our findings can contribute to immune-based patient stratification and facilitate the development of novel and durable (immune) therapeutic strategies in MM.
Collapse
Affiliation(s)
- Emma Verheye
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Translational Oncology Research Center, Lab of Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center of Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Daliya Kancheva
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center of Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Hatice Satilmis
- Translational Oncology Research Center, Lab of Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Niels Vandewalle
- Translational Oncology Research Center, Lab of Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Rong Fan
- Translational Oncology Research Center, Lab of Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Pauline M R Bardet
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center of Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Emile J Clappaert
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium
- Lab of Cellular and Molecular Immunology, Brussels Center of Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Kevin Verstaen
- VIB Single Cell Core, VIB, Ghent-Louvain, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Ann De Becker
- Translational Oncology Research Center, Lab of Hematology and Immunology, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Karin Vanderkerken
- Translational Oncology Research Center, Lab of Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Kim De Veirman
- Translational Oncology Research Center, Lab of Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
- Translational Oncology Research Center, Lab of Hematology and Immunology, Universitair Ziekenhuis Brussel (UZ Brussel), Laarbeeklaan 101, 1090, Brussels, Belgium.
| | - Damya Laoui
- Laboratory of Dendritic Cell Biology and Cancer Immunotherapy, VIB Center for Inflammation Research, Brussels, Belgium.
- Lab of Cellular and Molecular Immunology, Brussels Center of Immunology, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium.
| |
Collapse
|
10
|
Arzimanoglou I. The European Innovation Council supports innovative portfolios in health biotech. Nat Biotechnol 2024; 42:1488-1492. [PMID: 39402351 DOI: 10.1038/s41587-024-02416-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Affiliation(s)
- Iordanis Arzimanoglou
- Former Program Manager for Health and Biotechnology, EIC, Brussels, Belgium.
- Senior Advisor, Health Biotech, Brussels, Belgium.
| |
Collapse
|
11
|
Chen R, Shi Y, Fang N, Shao C, Huang H, Pan R, Xu Y, Wang M, Liu X, Xu K, Zhu R, Wang M. Bronchoalveolar lavage fluid analysis in patients with checkpoint inhibitor pneumonitis. Cancer Immunol Immunother 2024; 73:235. [PMID: 39271538 PMCID: PMC11399518 DOI: 10.1007/s00262-024-03834-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Checkpoint inhibitor pneumonitis (CIP) is a relatively uncommon but potentially life-threatening immune-related adverse event (irAE). Lung biopsies have not been commonly performed for CIP patients. Bronchoalveolar lavage fluid (BALF) analysis is a useful diagnostic approach for interstitial lung disease. However, BALF features were inconsistent across different studies. METHODS We retrospectively reviewed the medical records of 154 patients with pathologically confirmed malignancies and suffering from CIPs between July 2018 and December 2022. Patients who had bronchoalveolar lavage (BAL) data available were enrolled in our study. Patient clinical, laboratory, radiological and follow-up data were reviewed and analyzed. RESULTS The BALF differential cell count and lymphocyte subset analysis were performed for 42 CIP patients. There were 32 males (76.2%). The mean age at diagnosis of CIP was 62.0 ± 10.4 (range: 31-78) years. The median time to onset of CIP was 98.5 days after the start of immunotherapy. There were 18 patients (42.9%) with low-grade CIPs and 24 patients (57.1%) with high-grade CIPs. The mean lymphocyte percentage was 36.7 ± 22.5%. There were 34 (81%) CIP patients with a lymphocytic cellular pattern. The median ratio of CD3+CD4+/CD3+CD8+ lymphocytes was 0.5 (0.3, 1.0). The ratio was less than 1.0 for 31 CIP patients (73.8%). However, there was no significant difference in the BALF features between patients with low-grade CIPs and those with high-grade CIPs. CONCLUSIONS The CD3+CD8+ lymphocytosis pattern was the main inflammatory profile in the BALF of CIP patients in this cohort. Targeting CD3+CD8+ lymphocytes might be a treatment option for CIPs.
Collapse
Affiliation(s)
- Ruxuan Chen
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730, China
| | - Yujie Shi
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730, China
| | - Nan Fang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, #1 Xian Nong Tan Street, Beijing, 100050, China
| | - Chi Shao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730, China
| | - Hui Huang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730, China.
| | - Ruili Pan
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730, China
| | - Yan Xu
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730, China
| | - Mengqi Wang
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730, China
| | - Xiangning Liu
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730, China
| | - Kai Xu
- Department of Radiology, Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan Street, Dongcheng District, PekingBeijing, 100730, China
| | - Rui Zhu
- Department of Medical Records, Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan Street, Dongcheng District, PekingBeijing, 100730, China
| | - Mengzhao Wang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, #1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730, China
| |
Collapse
|
12
|
Langouo Fontsa M, Padonou F, Willard-Gallo K. Tumor-associated tertiary lymphoid structures in cancer: implications for immunotherapy. Expert Rev Clin Immunol 2024; 20:839-847. [PMID: 39007892 DOI: 10.1080/1744666x.2024.2380892] [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: 03/22/2023] [Accepted: 07/12/2024] [Indexed: 07/16/2024]
Abstract
INTRODUCTION Tertiary lymphoid structures (TLS) arise at chronic inflammatory sites where they function as miniature lymph nodes to generate immune responses, which can be beneficial or detrimental, in diseases as diverse as autoimmunity, chronic infections and cancer. A growing number of studies show that a TLS presence in tumors from cancer patients treated with immune checkpoint inhibitors is closely linked with improved clinical outcomes. TLS may foster the generation of specific anti-tumor immune responses and immunological memory that recognizes a patient's own tumor. Due to repeated rounds of chronic inflammation, some tumor-associated TLS may be immunologically inactive, with immune checkpoint inhibitors functioning to revitalize them through pathway activation. AREAS COVERED This review summarizes work on TLS and how they mediate immune responses in human tumors. We also explore TLS as potential prognostic and predictive biomarkers for immunotherapy. EXPERT OPINION The presence of TLS in human tumors has been linked with a better clinical prognosis, response to treatment(s) and overall survival. TLS provide a structured microenvironment for the activation, expansion and maturation of immune cells at the tumor site. These activities can enhance the efficacy of immunotherapeutic treatments such as checkpoint inhibitors and cancer vaccines by revitalizing local anti-tumor immunity.
Collapse
Affiliation(s)
- Mireille Langouo Fontsa
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Francine Padonou
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Karen Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
13
|
Glehr G, Riquelme P, Kronenberg K, Lohmayer R, López-Madrona VJ, Kapinsky M, Schlitt HJ, Geissler EK, Spang R, Haferkamp S, Hutchinson JA. Restricting datasets to classifiable samples augments discovery of immune disease biomarkers. Nat Commun 2024; 15:5417. [PMID: 38926389 PMCID: PMC11208602 DOI: 10.1038/s41467-024-49094-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 05/14/2024] [Indexed: 06/28/2024] Open
Abstract
Immunological diseases are typically heterogeneous in clinical presentation, severity and response to therapy. Biomarkers of immune diseases often reflect this variability, especially compared to their regulated behaviour in health. This leads to a common difficulty that frustrates biomarker discovery and interpretation - namely, unequal dispersion of immune disease biomarker expression between patient classes necessarily limits a biomarker's informative range. To solve this problem, we introduce dataset restriction, a procedure that splits datasets into classifiable and unclassifiable samples. Applied to synthetic flow cytometry data, restriction identifies biomarkers that are otherwise disregarded. In advanced melanoma, restriction finds biomarkers of immune-related adverse event risk after immunotherapy and enables us to build multivariate models that accurately predict immunotherapy-related hepatitis. Hence, dataset restriction augments discovery of immune disease biomarkers, increases predictive certainty for classifiable samples and improves multivariate models incorporating biomarkers with a limited informative range. This principle can be directly extended to any classification task.
Collapse
Affiliation(s)
- Gunther Glehr
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Paloma Riquelme
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | | | - Robert Lohmayer
- Algorithmic Bioinformatics Research Group, Leibniz Institute for Immunotherapy, Regensburg, Germany
| | | | | | - Hans J Schlitt
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Rainer Spang
- Department of Statistical Bioinformatics, University of Regensburg, Regensburg, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | - James A Hutchinson
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany.
| |
Collapse
|
14
|
Choudhury SR, Byrum SD, Blossom SJ. Trichloroethylene metabolite modulates DNA methylation-dependent gene expression in Th1-polarized CD4+ T cells from autoimmune-prone mice. Toxicol Sci 2024; 199:289-300. [PMID: 38518092 PMCID: PMC11131021 DOI: 10.1093/toxsci/kfae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024] Open
Abstract
Trichloroethylene (TCE) is an industrial solvent and widespread environmental contaminant associated with CD4+ T-cell activation and autoimmune disease. Prior studies showed that exposure to TCE in the drinking water of autoimmune-prone mice expanded effector/memory CD4+ T cells with an interferon-γ (IFN-γ)-secreting Th1-like phenotype. However, very little is known how TCE exposure skews CD4+ T cells towards this pro-inflammatory Th1 subset. As observed previously, TCE exposure was associated with hypermethylation of regions of the genome related to transcriptional repression in purified effector/memory CD4 T cells. We hypothesized that TCE modulates transcriptional and/or epigenetic programming of CD4+ T cells as they differentiate from a naive to effector phenotype. In the current study, purified naive CD4 T cells from both male and female autoimmune-prone MRL/MpJ mice were activated ex vivo and polarized towards a Th1 subset for 4 days in the presence or absence of the oxidative metabolite of TCE, trichloroacetaldehyde hydrate (TCAH) in vitro. An RNA-seq assessment and reduced representation bisulfite sequencing for DNA methylation were conducted on Th1 cells or activated, non-polarized cells. The results demonstrated TCAH's ability to regulate key genes involved in the immune response and autoimmunity, including Ifng, by altering the level of DNA methylation at the gene promoter. Intriguing sex differences were observed and for the most part, the effects were more robust in females compared to males. In conclusion, TCE via TCAH epigenetically regulates gene expression in CD4+ T cells. These results may have implications for mechanistic understanding or future therapeutics for autoimmunity.
Collapse
Affiliation(s)
- Samrat Roy Choudhury
- Division of Hematology/Oncology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72202, USA
- Arkansas Children’s Research Institute, Department of Pediatrics, Little Rock, Arkansas 72202, USA
| | - Stephanie D Byrum
- Arkansas Children’s Research Institute, Department of Pediatrics, Little Rock, Arkansas 72202, USA
- Department of Biochemistry & Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, USA
| | - Sarah J Blossom
- Department of Pharmaceutical Sciences, University of New Mexico, Albuquerque, New Mexico 87131, USA
| |
Collapse
|
15
|
Doan AE, Mueller KP, Chen AY, Rouin GT, Chen Y, Daniel B, Lattin J, Markovska M, Mozarsky B, Arias-Umana J, Hapke R, Jung IY, Wang A, Xu P, Klysz D, Zuern G, Bashti M, Quinn PJ, Miao Z, Sandor K, Zhang W, Chen GM, Ryu F, Logun M, Hall J, Tan K, Grupp SA, McClory SE, Lareau CA, Fraietta JA, Sotillo E, Satpathy AT, Mackall CL, Weber EW. FOXO1 is a master regulator of memory programming in CAR T cells. Nature 2024; 629:211-218. [PMID: 38600391 PMCID: PMC11062920 DOI: 10.1038/s41586-024-07300-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 03/12/2024] [Indexed: 04/12/2024]
Abstract
A major limitation of chimeric antigen receptor (CAR) T cell therapies is the poor persistence of these cells in vivo1. The expression of memory-associated genes in CAR T cells is linked to their long-term persistence in patients and clinical efficacy2-6, suggesting that memory programs may underpin durable CAR T cell function. Here we show that the transcription factor FOXO1 is responsible for promoting memory and restraining exhaustion in human CAR T cells. Pharmacological inhibition or gene editing of endogenous FOXO1 diminished the expression of memory-associated genes, promoted an exhaustion-like phenotype and impaired the antitumour activity of CAR T cells. Overexpression of FOXO1 induced a gene-expression program consistent with T cell memory and increased chromatin accessibility at FOXO1-binding motifs. CAR T cells that overexpressed FOXO1 retained their function, memory potential and metabolic fitness in settings of chronic stimulation, and exhibited enhanced persistence and tumour control in vivo. By contrast, overexpression of TCF1 (encoded by TCF7) did not enforce canonical memory programs or enhance the potency of CAR T cells. Notably, FOXO1 activity correlated with positive clinical outcomes of patients treated with CAR T cells or tumour-infiltrating lymphocytes, underscoring the clinical relevance of FOXO1 in cancer immunotherapy. Our results show that overexpressing FOXO1 can increase the antitumour activity of human CAR T cells, and highlight memory reprogramming as a broadly applicable approach for optimizing therapeutic T cell states.
Collapse
Affiliation(s)
- Alexander E Doan
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Katherine P Mueller
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andy Y Chen
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Geoffrey T Rouin
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yingshi Chen
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bence Daniel
- Department of Pathology, Stanford University, Stanford, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Genentech, South San Francisco, CA, USA
| | - John Lattin
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Martina Markovska
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brett Mozarsky
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jose Arias-Umana
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robert Hapke
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - In-Young Jung
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alice Wang
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peng Xu
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Dorota Klysz
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Gabrielle Zuern
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Malek Bashti
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Patrick J Quinn
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhuang Miao
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Katalin Sandor
- Department of Pathology, Stanford University, Stanford, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Wenxi Zhang
- Department of Pathology, Stanford University, Stanford, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Gregory M Chen
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Faith Ryu
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Meghan Logun
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Junior Hall
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kai Tan
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephan A Grupp
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Susan E McClory
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Caleb A Lareau
- Department of Pathology, Stanford University, Stanford, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Joseph A Fraietta
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elena Sotillo
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University, Stanford, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
- Department of Medicine, Stanford University, Stanford, CA, USA.
| | - Evan W Weber
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| |
Collapse
|
16
|
von Itzstein MS, Yang Y, Wang Y, Hsiehchen D, Sheffield TY, Fattah F, Popat V, Ahmed M, Homsi J, Dowell JE, Rashdan S, Lohrey J, Hammers HJ, Hughes RS, Wang T, Xie Y, Gerber DE. Highly variable timing renders immunotherapy efficacy and toxicity impractical biomarkers of one another in clinical practice. Front Immunol 2024; 15:1351739. [PMID: 38690281 PMCID: PMC11058939 DOI: 10.3389/fimmu.2024.1351739] [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: 12/06/2023] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Background A useful clinical biomarker requires not only association but also a consistent temporal relationship. For instance, chemotherapy-induced neutropenia and epidermal growth-factor inhibitor-related acneiform rash both occur within weeks of treatment initiation, thereby providing information prior to efficacy assessment. Although immune checkpoint inhibitor (ICI)-associated immune-related adverse events (irAE) have been associated with therapeutic benefit, irAE may have delayed and highly variable onset. To determine whether ICI efficacy and irAE could serve as clinically useful biomarkers for predicting each other, we determined the temporal relationship between initial efficacy assessment and irAE onset in a diverse population treated with ICI. Methods Using two-sided Fisher exact and Cochran-Armitage tests, we determined the relative timing of initial efficacy assessment and irAE occurrence in a cohort of 155 ICI-treated patients (median age 68 years, 40% women). Results Initial efficacy assessment was performed a median of 50 days [interquartile range (IQR) 39-59 days] after ICI initiation; median time to any irAE was 77 days (IQR 28-145 days) after ICI initiation. Median time to first irAE was 42 days (IQR 20-88 days). Overall, 58% of any irAE and 47% of first irAE occurred after initial efficacy assessment. For clinically significant (grade ≥2) irAE, 60% of any and 53% of first occurred after initial efficacy assessment. The likelihood of any future irAE did not differ according to response (45% for complete or partial response vs. 47% for other cases; P=1). In landmark analyses controlling for clinical and toxicity follow-up, patients demonstrating greater tumor shrinkage at initial efficacy assessment were more likely to develop future grade ≥2 (P=0.05) and multi-organ (P=0.02) irAE. Conclusions In contrast to that seen with chemotherapy and molecularly targeted therapies, the temporal relationship between ICI efficacy and toxicity is complex and bidirectional. In practice, neither parameter can be routinely relied on as a clinical biomarker to predict the other.
Collapse
Affiliation(s)
- Mitchell S. von Itzstein
- Department of Internal Medicine (Division of Hematology-Oncology), Dallas, TX, United States
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yuqiu Yang
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yiqing Wang
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David Hsiehchen
- Department of Internal Medicine (Division of Hematology-Oncology), Dallas, TX, United States
| | - Thomas Y. Sheffield
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Farjana Fattah
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Vinita Popat
- School of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Murtaza Ahmed
- School of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jade Homsi
- Department of Internal Medicine (Division of Hematology-Oncology), Dallas, TX, United States
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jonathan E. Dowell
- Department of Internal Medicine (Division of Hematology-Oncology), Dallas, TX, United States
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Sawsan Rashdan
- Department of Internal Medicine (Division of Hematology-Oncology), Dallas, TX, United States
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Jay Lohrey
- Department of Internal Medicine (Division of Hematology-Oncology), Dallas, TX, United States
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Hans J. Hammers
- Department of Internal Medicine (Division of Hematology-Oncology), Dallas, TX, United States
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Randall S. Hughes
- Department of Internal Medicine (Division of Hematology-Oncology), Dallas, TX, United States
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Tao Wang
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yang Xie
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David E. Gerber
- Department of Internal Medicine (Division of Hematology-Oncology), Dallas, TX, United States
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
17
|
Jani Y, Jansen CS, Gerke MB, Bilen MA. Established and emerging biomarkers of immunotherapy in renal cell carcinoma. Immunotherapy 2024; 16:405-426. [PMID: 38264827 PMCID: PMC11913054 DOI: 10.2217/imt-2023-0267] [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: 10/17/2023] [Accepted: 01/08/2024] [Indexed: 01/25/2024] Open
Abstract
Immunotherapies, such as immune checkpoint inhibitors, have heralded impressive progress for patient care in renal cell carcinoma (RCC). Despite this success, some patients' disease fails to respond, and other patients experience significant side effects. Thus, development of biomarkers is needed to ensure that patients can be selected to maximize benefit from immunotherapies. Improving clinicians' ability to predict which patients will respond to immunotherapy and which are most at risk of adverse events - namely through clinical biomarkers - is indispensable for patient safety and therapeutic efficacy. Accordingly, an evolving suite of therapeutic biomarkers continues to be investigated. This review discusses biomarkers for immunotherapy in RCC, highlighting current practices and emerging innovations, aiming to contribute to improved outcomes for patients with RCC.
Collapse
Affiliation(s)
- Yash Jani
- Mercer University, Macon, GA31207, USA
| | - Caroline S Jansen
- Emory University School of Medicine, Atlanta, GA30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA30322, USA
| | - Margo B Gerke
- Emory University School of Medicine, Atlanta, GA30322, USA
| | - Mehmet Asim Bilen
- Winship Cancer Institute of Emory University, Atlanta, GA30322, USA
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Atlanta, GA30322, USA
| |
Collapse
|
18
|
Suijkerbuijk KPM, van Eijs MJM, van Wijk F, Eggermont AMM. Clinical and translational attributes of immune-related adverse events. NATURE CANCER 2024; 5:557-571. [PMID: 38360861 DOI: 10.1038/s43018-024-00730-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/12/2024] [Indexed: 02/17/2024]
Abstract
With immune checkpoint inhibitors (ICIs) becoming the mainstay of treatment for many cancers, managing their immune-related adverse events (irAEs) has become an important part of oncological care. This Review covers the clinical presentation of irAEs and crucial aspects of reversibility, fatality and long-term sequelae, with special attention to irAEs in specific patient populations, such as those with autoimmune diseases. In addition, the genetic basis of irAEs, along with cellular and humoral responses to ICI therapy, are discussed. Detrimental effects of empirically used high-dose steroids and second-line immunosuppression, including impaired ICI effectiveness, call for more tailored irAE-treatment strategies. We discuss open therapeutic challenges and propose potential avenues to accelerate personalized management strategies and optimize outcomes.
Collapse
Affiliation(s)
- Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| | - Mick J M van Eijs
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Alexander M M Eggermont
- University Medical Center Utrecht and Princess Máxima Center, Utrecht, the Netherlands
- Comprehensive Cancer Center Munich of the Technical University of Munich and the Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
19
|
Lin X, Xie M, Yao J, Ma X, Qin L, Zhang X, Song J, Bao X, Zhang X, Zhang Y, Liu Y, Han W, Liang Y, Jing Y, Xue X. Immune-related adverse events in non-small cell lung cancer: Occurrence, mechanisms and therapeutic strategies. Clin Transl Med 2024; 14:e1613. [PMID: 38451000 PMCID: PMC10918746 DOI: 10.1002/ctm2.1613] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 03/08/2024] Open
Abstract
The emergence of immune checkpoint inhibitors (ICIs) has heralded a transformative era in the therapeutic landscape of non-small cell lung cancer (NSCLC). While ICIs have demonstrated clinical efficacy in a portion of patients with NSCLC, these treatments concurrently precipitate a spectrum of immune-related adverse events (irAEs), encompassing mild to severe manifestations, collectively posing a risk of significant organ damage. Consequently, there exists an imperative to augment our comprehension of the pathophysiological underpinnings of irAEs and to formulate more efficacious preventive and ameliorative strategies. In this comprehensive review, we delineate the clinical presentation of organ-specific irAEs in patients with NSCLC and provide an in-depth analysis of recent advancements in understanding the mechanisms driving ICI-induced toxicity. Furthermore, we discuss potential strategies and targets for ameliorating these irAEs. Ultimately, this review aims to furnish valuable insights to guide further research endeavours in the context of irAEs in NSCLC patients.
Collapse
Affiliation(s)
- Xuwen Lin
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Mei Xie
- Department of Respiratory and Critical CareChinese PLA General HospitalBeijingChina
| | - Jie Yao
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Xidong Ma
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Lin Qin
- Department of Endoscopic Diagnosis and TreatmentTuberculosis and Thoracic Tumor InstituteBeijing Chest HospitalCapital Medical UniversityBeijingChina
| | - Xu‐Mei Zhang
- Department of PathologyAffiliated Hospital of Weifang Medical UniversityWeifangShandongChina
| | - Jialin Song
- Department of Respiratory and Critical CareShandong Second Medical UniversityShandongChina
| | - Xinyu Bao
- Department of Respiratory and Critical CareShandong Second Medical UniversityShandongChina
| | - Xin Zhang
- Department of Respiratory and Critical CareShandong Second Medical UniversityShandongChina
| | - Yinguang Zhang
- Department of Thoracic SurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Yiming Liu
- Department of Thoracic SurgeryChinese PLA General HospitalBeijingChina
| | - Wenya Han
- Department of Respiratory and Critical CareTaihe HospitalHubei University of MedicineShiyanChina
| | - Yiran Liang
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
| | - Ying Jing
- Center for Intelligent MedicineGreater Bay Area Institute of Precision Medicine (Guangzhou)School of Life SciencesFudan UniversityGuangzhouGuangdongChina
| | - Xinying Xue
- Department of Respiratory and Critical CareEmergency and Critical Care Medical CenterBeijing Shijitan HospitalCapital Medical UniversityBeijingChina
- Department of Respiratory and Critical CareShandong Second Medical UniversityShandongChina
| |
Collapse
|
20
|
Chen G, Xu W, Long Z, Chong Y, Lin B, Jie Y. Single-cell Technologies Provide Novel Insights into Liver Physiology and Pathology. J Clin Transl Hepatol 2024; 12:79-90. [PMID: 38250462 PMCID: PMC10794276 DOI: 10.14218/jcth.2023.00224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/25/2023] [Accepted: 07/12/2023] [Indexed: 01/23/2024] Open
Abstract
The liver is the largest glandular organ in the body and has a unique distribution of cells and biomolecules. However, the treatment outcome of end-stage liver disease is extremely poor. Single-cell sequencing is a new advanced and powerful technique for identifying rare cell populations and biomolecules by analyzing the characteristics of gene expression between individual cells. These cells and biomolecules might be used as potential targets for immunotherapy of liver diseases and contribute to the development of precise individualized treatment. Compared to whole-tissue RNA sequencing, single-cell RNA sequencing (scRNA-seq) or other single-cell histological techniques have solved the problem of cell population heterogeneity and characterize molecular changes associated with liver diseases with higher accuracy and resolution. In this review, we comprehensively summarized single-cell approaches including transcriptomic, spatial transcriptomic, immunomic, proteomic, epigenomic, and multiomic technologies, and described their application in liver physiology and pathology. We also discussed advanced techniques and recent studies in the field of single-cell; our review might provide new insights into the pathophysiological mechanisms of the liver to achieve precise and individualized treatment of liver diseases.
Collapse
Affiliation(s)
| | | | - Zhicong Long
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yutian Chong
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Bingliang Lin
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yusheng Jie
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
21
|
Straube J, Bukhari S, Lerrer S, Winchester RJ, Gartshteyn Y, Henick BS, Dragovich MA, Mor A. PD-1 signaling uncovers a pathogenic subset of T cells in inflammatory arthritis. Arthritis Res Ther 2024; 26:32. [PMID: 38254179 PMCID: PMC10801937 DOI: 10.1186/s13075-023-03259-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND PD-1 is an immune checkpoint on T cells, and interventions to block this receptor result in T cell activation and enhanced immune response to tumors and pathogens. Reciprocally, despite a decade of research, approaches to treat autoimmunity with PD-1 agonists have only had limited successful. To resolve this, new methods must be developed to augment PD-1 function beyond engaging the receptor. METHODS We conducted a flow cytometry analysis of T cells isolated from the peripheral blood and synovial fluid of patients with rheumatoid arthritis. In addition, we performed a genome-wide CRISPR/Cas9 screen to identify genes associated with PD-1 signaling. We further analyzed genes involved in PD-1 signaling using publicly available bulk and single-cell RNA sequencing datasets. RESULTS Our screen confirmed known regulators in proximal PD-1 signaling and, importantly, identified an additional 1112 unique genes related to PD-1 ability to inhibit T cell functions. These genes were strongly associated with the response of cancer patients to PD-1 blockades and with high tumor immune dysfunction and exclusion scores, confirming their role downstream of PD-1. Functional annotation revealed that the most significant genes uncovered were those associated with known immune regulation processes. Remarkably, these genes were considerably downregulated in T cells isolated from patients with inflammatory arthritis, supporting their overall inhibitory functions. A study of rheumatoid arthritis single-cell RNA sequencing data demonstrated that five genes, KLRG1, CRTAM, SLAMF7, PTPN2, and KLRD1, were downregulated in activated and effector T cells isolated from synovial fluids. Backgating these genes to canonical cytotoxic T cell signatures revealed PD-1+ HLA-DRHIGH KLRG1LOW T cells as a novel inflammatory subset of T cells. CONCLUSIONS We concluded that PD-1+ HLA-DRHIGH KLRG1LOW T cells are a potential target for future PD-1 agonists to treat inflammatory diseases. Our study uncovers new genes associated with PD-1 downstream functions and, therefore, provides a comprehensive resource for additional studies that are much needed to characterize the role of PD-1 in the synovial subset of T cells.
Collapse
Affiliation(s)
- Johanna Straube
- Columbia Center for Translational Immunology, Columbia University Medical Center, 650 W 168 St. BB-1701F, New York, NY, 10032, USA
- Institute of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06099, Halle (Saale), Germany
| | - Shoiab Bukhari
- Columbia Center for Translational Immunology, Columbia University Medical Center, 650 W 168 St. BB-1701F, New York, NY, 10032, USA
| | - Shalom Lerrer
- Columbia Center for Translational Immunology, Columbia University Medical Center, 650 W 168 St. BB-1701F, New York, NY, 10032, USA
| | - Robert J Winchester
- Columbia Center for Translational Immunology, Columbia University Medical Center, 650 W 168 St. BB-1701F, New York, NY, 10032, USA
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Yevgeniya Gartshteyn
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| | - Brian S Henick
- Herbert Irving Cancer Center, Columbia University Medical Center, New York, NY, 10032, USA
| | - Matthew A Dragovich
- Columbia Center for Translational Immunology, Columbia University Medical Center, 650 W 168 St. BB-1701F, New York, NY, 10032, USA
| | - Adam Mor
- Columbia Center for Translational Immunology, Columbia University Medical Center, 650 W 168 St. BB-1701F, New York, NY, 10032, USA.
- Division of Rheumatology, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA.
- Herbert Irving Cancer Center, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
22
|
Kovacsovics-Bankowski M, Sweere JM, Healy CP, Sigal N, Cheng LC, Chronister WD, Evans SA, Marsiglio J, Gibson B, Swami U, Erickson-Wayman A, McPherson JP, Derose YS, Eliason AL, Medina CO, Srinivasan R, Spitzer MH, Nguyen N, Hyngstrom J, Hu-Lieskovan S. Lower frequencies of circulating suppressive regulatory T cells and higher frequencies of CD4 + naïve T cells at baseline are associated with severe immune-related adverse events in immune checkpoint inhibitor-treated melanoma. J Immunother Cancer 2024; 12:e008056. [PMID: 38233101 PMCID: PMC10806651 DOI: 10.1136/jitc-2023-008056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 01/19/2024] Open
Abstract
BACKGROUND Immune-related adverse events (irAEs) are major barriers of clinical management and further development of immune checkpoint inhibitors (ICIs) for cancer therapy. Therefore, biomarkers associated with the onset of severe irAEs are needed. In this study, we aimed to identify immune features detectable in peripheral blood and associated with the development of severe irAEs that required clinical intervention. METHODS We used a 43-marker mass cytometry panel to characterize peripheral blood mononuclear cells from 28 unique patients with melanoma across 29 lines of ICI therapy before treatment (baseline), before the onset of irAEs (pre-irAE) and at the peak of irAEs (irAE-max). In the 29 lines of ICI therapy, 18 resulted in severe irAEs and 11 did not. RESULTS Unsupervised and gated population analysis showed that patients with severe irAEs had a higher frequency of CD4+ naïve T cells and lower frequency of CD16+ natural killer (NK) cells at all time points. Gated population analysis additionally showed that patients with severe irAEs had fewer T cell immunoreceptor with Ig and ITIM domain (TIGIT+) regulatory T cells at baseline and more activated CD38+ CD4+ central memory T cells (TCM) and CD39+ and Human Leukocyte Antigen-DR Isotype (HLA-DR)+ CD8+ TCM at peak of irAEs. The differentiating immune features at baseline were predominantly seen in patients with gastrointestinal and cutaneous irAEs and type 1 diabetes. Higher frequencies of CD4+ naïve T cells and lower frequencies of CD16+ NK cells were also associated with clinical benefit to ICI therapy. CONCLUSIONS This study demonstrates that high-dimensional immune profiling can reveal novel blood-based immune signatures associated with risk and mechanism of severe irAEs. Development of severe irAEs in melanoma could be the result of reduced immune inhibitory capacity pre-ICI treatment, resulting in more activated TCM cells after treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - John Marsiglio
- The University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Berit Gibson
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Umang Swami
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Alyssa Erickson-Wayman
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jordan P McPherson
- Department of Pharmacy, Huntsman Cancer Institute Cancer Hospital, Salt Lake City, Utah, USA
| | - Yoko S Derose
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | | | | | | | - Matthew H Spitzer
- Teiko.bio Inc, Salt Lake City, Utah, USA
- Department of Otolaryngology-Head and Neck Cancer, University of California San Francisco, San Francisco, California, USA
| | | | - John Hyngstrom
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Siwen Hu-Lieskovan
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
23
|
Cina ML, Venegas J, Young A. Stocking the toolbox-Using preclinical models to understand the development and treatment of immune checkpoint inhibitor-induced immune-related adverse events. Immunol Rev 2023; 318:110-137. [PMID: 37565407 PMCID: PMC10529261 DOI: 10.1111/imr.13250] [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: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 08/12/2023]
Abstract
Cancer patients treated with immune checkpoint inhibitors (ICIs) are susceptible to a broad and variable array of immune-related adverse events (irAEs). With increasing clinical use of ICIs, defining the mechanism for irAE development is more critical than ever. However, it currently remains challenging to predict when these irAEs occur and which organ may be affected, and for many of the more severe irAEs, inaccessibility to the tissue site hampers mechanistic insight. This lack of understanding of irAE development in the clinical setting emphasizes the need for greater use of preclinical models that allow for improved prediction of biomarkers for ICI-initiated irAEs or that validate treatment options that inhibit irAEs without hampering the anti-tumor immune response. Here, we discuss the utility of preclinical models, ranging from exploring databases to in vivo animal models, focusing on where they are most useful and where they could be improved.
Collapse
Affiliation(s)
- Morgan L Cina
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jessica Venegas
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Arabella Young
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| |
Collapse
|
24
|
Earland N, Zhang W, Usmani A, Nene A, Bacchiocchi A, Chen DY, Sznol M, Halaban R, Chaudhuri AA, Newman AM. CD4 T cells and toxicity from immune checkpoint blockade. Immunol Rev 2023; 318:96-109. [PMID: 37491734 PMCID: PMC10838135 DOI: 10.1111/imr.13248] [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: 04/15/2023] [Accepted: 06/27/2023] [Indexed: 07/27/2023]
Abstract
Immune-related toxicities, otherwise known as immune-related adverse events (irAEs), occur in a substantial fraction of cancer patients treated with immune checkpoint inhibitors (ICIs). Ranging from asymptomatic to life-threatening, ICI-induced irAEs can result in hospital admission, high-dose corticosteroid treatment, ICI discontinuation, and in some cases, death. A deeper understanding of the factors underpinning severe irAE development will be essential for improved irAE prediction and prevention, toward maximizing the benefits and safety profiles of ICIs. In recent work, we applied mass cytometry, single-cell RNA sequencing, single-cell V(D)J sequencing, bulk RNA sequencing, and bulk T-cell receptor (TCR) sequencing to identify pretreatment determinants of severe irAE development in patients with advanced melanoma. Across 71 patients separated into three cohorts, we found that two baseline features in circulation-elevated activated CD4 effector memory T-cell abundance and TCR diversity-are associated with severe irAE development, independent of the affected organ system within 3 months of ICI treatment initiation. Here, we provide an extended perspective on this work, synthesize and discuss related literature, and summarize practical considerations for clinical translation. Collectively, these findings lay a foundation for data-driven and mechanistic insights into irAE development, with the potential to reduce ICI morbidity and mortality in the future.
Collapse
Affiliation(s)
- Noah Earland
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Wubing Zhang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Abul Usmani
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Antonella Bacchiocchi
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - David Y. Chen
- Division of Dermatology, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Mario Sznol
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Medicine, Division of Medical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Ruth Halaban
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Aadel A. Chaudhuri
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Aaron M. Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
25
|
Singh N, Hocking AM, Buckner JH. Immune-related adverse events after immune check point inhibitors: Understanding the intersection with autoimmunity. Immunol Rev 2023; 318:81-88. [PMID: 37493210 DOI: 10.1111/imr.13247] [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: 03/31/2023] [Accepted: 06/28/2023] [Indexed: 07/27/2023]
Abstract
Immune checkpoint inhibitor therapies act through blockade of inhibitory molecules involved in the regulation of T cells, thus releasing tumor specific T cells to destroy their tumor targets. However, immune checkpoint inhibitors (ICI) can also lead to a breach in self-tolerance resulting in immune-related adverse events (irAEs) that include tissue-specific autoimmunity. This review addresses the question of whether the mechanisms that drive ICI-induced irAEs are shared or distinct with those driving spontaneous autoimmunity, focusing on ICI-induced diabetes, ICI-induced arthritis, and ICI-induced thyroiditis due to the wealth of knowledge about the development of autoimmunity in type 1 diabetes, rheumatoid arthritis, and Hashimoto's thyroiditis. It reviews current knowledge about role of genetics and autoantibodies in the development of ICI-induced irAEs and presents new studies utilizing single-cell omics approaches to identify T-cell signatures associated with ICI-induced irAEs. Collectively, these studies indicate that there are similarities and differences between ICI-induced irAEs and autoimmune disease and that studying them in parallel will provide important insight into the mechanisms critical for maintaining immune tolerance.
Collapse
Affiliation(s)
- Namrata Singh
- Division of Rheumatology, University of Washington, Seattle, Washington, USA
| | - Anne M Hocking
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | - Jane H Buckner
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| |
Collapse
|
26
|
Curkovic NB, Johnson DB. Updates in toxicities associated with immune checkpoint inhibitors. Expert Rev Clin Immunol 2023; 19:1117-1129. [PMID: 37276071 PMCID: PMC10527235 DOI: 10.1080/1744666x.2023.2221434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/31/2023] [Indexed: 06/07/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have become a pillar of treatment for numerous cancers with increasing use in combination with other ICIs and in earlier stages of disease treatment. Although effective, ICI use is accompanied by a milieu of potentially bothersome or even life-threatening toxicities known as immune-related adverse events (irAEs), necessitating careful monitoring and early intervention. AREAS COVERED In this review, we provide an overview of recent advances surrounding toxicity pathophysiology and treatment in the context of relevant organ systems. An emphasis on current treatments by toxicity, as well as updates on steroid-refractory toxicities, chronic toxicities, and biomarkers will be a focus of this update on the current understanding of irAEs. EXPERT OPINION ICI toxicities are a major limitation on the deployment of multi-agent ICI regimens and are thus a major priority to understand, treat, and prevent. Recent developments have led to greater understanding of the pathophysiology of these events, which may lead to improved prevention or mitigation strategies. Further, early studies have also suggested steroid-sparing approaches that may be useful. Ultimately, preventing and managing irAEs will be a key goal toward successful ICI treatment across a broader range of patients with cancer.
Collapse
Affiliation(s)
| | - Douglas B. Johnson
- Department of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
27
|
Cao T, Zhou X, Wu X, Zou Y. Cutaneous immune-related adverse events to immune checkpoint inhibitors: from underlying immunological mechanisms to multi-omics prediction. Front Immunol 2023; 14:1207544. [PMID: 37497220 PMCID: PMC10368482 DOI: 10.3389/fimmu.2023.1207544] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/05/2023] [Indexed: 07/28/2023] Open
Abstract
The development of immune checkpoint inhibitors (ICIs) has dramatically altered the landscape of therapy for multiple malignancies, including urothelial carcinoma, non-small cell lung cancer, melanoma and gastric cancer. As part of their anti-tumor properties, ICIs can enhance susceptibility to inflammatory side effects known as immune-related adverse events (irAEs), in which the skin is one of the most commonly and rapidly affected organs. Although numerous questions still remain unanswered, multi-omics technologies have shed light into immunological mechanisms, as well as the correlation between ICI-induced activation of immune systems and the incidence of cirAE (cutaneous irAEs). Therefore, we reviewed integrated biological layers of omics studies combined with clinical data for the prediction biomarkers of cirAEs based on skin pathogenesis. Here, we provide an overview of a spectrum of dermatological irAEs, discuss the pathogenesis of this "off-tumor toxicity" during ICI treatment, and summarize recently investigated biomarkers that may have predictive value for cirAEs via multi-omics approach. Finally, we demonstrate the prognostic significance of cirAEs for immune checkpoint blockades.
Collapse
|
28
|
Yang J, Chen Y, Han L. Dynamic immune signatures as biomarkers for irAEs. Trends Cancer 2023; 9:379-380. [PMID: 36941187 DOI: 10.1016/j.trecan.2023.03.004] [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: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/23/2023]
Abstract
Identifying predictive biomarkers of immune-related adverse events (irAEs) is crucial to maximize the benefits for patients with cancer treated with immune checkpoint inhibitors (ICIs). In a recent study published in Med,Nuñez et al. utilized multi-omics approaches and identified blood immune signatures that have the potential to predict the development of autoimmune toxicity.
Collapse
Affiliation(s)
- Jingwen Yang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Yamei Chen
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Leng Han
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA; Department of Translational Medical Sciences, College of Medicine, Texas A&M University, Houston, TX, USA.
| |
Collapse
|
29
|
Müller B, Bärenwaldt A, Herzig P, Zippelius A, Maul LV, Hess V, König D, Läubli H. Changes of peripheral T cell subsets in melanoma patients with immune-related adverse events. Front Immunol 2023; 14:1125111. [PMID: 37122748 PMCID: PMC10130408 DOI: 10.3389/fimmu.2023.1125111] [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: 12/15/2022] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Immunotherapies have improved the prognosis of many cancer patients including patients with advanced melanoma. Immune checkpoint receptors including CTLA-4 and PD-1 have been established as main therapeutic targets for immunotherapy of melanoma. Although monotherapy is effective in melanoma patients, a dual therapy approach has been shown to be most effective. Dual checkpoint blockade, however, increases substantially the risk for immune-related adverse events (irAEs). Methods In this study, we characterized peripheral immune cell subsets in patients with anti-PD-1 monotherapy and with dual immune receptors blockade targeting PD-1 and CTLA-4. Results We found differences in peripheral T cells between patients who developed severe immune-related side effects and patients with mild irAEs. We identified several mainly changes in CD8+ T cell subsets in patients with severe irAE under dual PD-1 and CTLA-4 blockade. Discussion This work suggests that peripheral immune cell dynamics could be associated with severe immune-related side effects in patients receiving immune checkpoint inhibitors. These changes could be used as future biomarkers in early diagnosis of irAEs.
Collapse
Affiliation(s)
- Benjamin Müller
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Anne Bärenwaldt
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Petra Herzig
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Alfred Zippelius
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Oncology, University Hospital Basel, Basel, Switzerland
| | - Lara Valeska Maul
- Department of Dermatology, University Hospital Basel, Basel, Switzerland
| | - Viviane Hess
- Division of Oncology, University Hospital Basel, Basel, Switzerland
| | - David König
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Oncology, University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
30
|
Distinct T cell sub-clusters may serve as biomarkers for immune related adverse events. Cell Rep Med 2023; 4:100902. [PMID: 36652911 PMCID: PMC9873920 DOI: 10.1016/j.xcrm.2022.100902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Identifying biomarkers of irAEs is the prerequisite for maximizing clinical benefits of patients treated by immune checkpoint inhibitors. Bukhari et al.1 identified significant associations between different peripheral T cell sub-clusters and arthritis, pneumonitis, and thyroiditis.
Collapse
|