1
|
Fan G, Zuo S, Wang Z, Zhang S, Liu L, Luo H, Xie Y, Zhang Y, Li D. Targeting of the IL-33/Wnt axis restricts breast cancer stemness and metastasis. Sci Rep 2025; 15:18172. [PMID: 40414980 DOI: 10.1038/s41598-025-03260-9] [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: 02/20/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Interleukin-33 (IL-33) plays multifaceted roles in tumor progression, but its autocrine regulation of breast cancer stemness and metastasis via the Wnt pathway remains unclear. Here, we investigated the IL-33/ST2 axis in breast cancer using CRISPR/Cas9, single-cell RNA sequencing, and murine models (orthotopic 4T1 and spontaneous MMTV-PyMT). Elevated IL-33 levels correlated with aggressive subtypes and poor prognosis. IL-33 overexpression enhanced proliferation, migration, and cancer stem cell (CSC) marker expression (CD44, ALDH1) in 4T1 and MDA-MB-231 cells, whereas ST2 knockdown via CRISPR or adeno-associated virus (AAV) attenuated tumor growth and metastasis in vivo, reducing CSC frequency. Mechanistically, IL-33 activated Wnt/β-catenin signaling to promote stemness, which was reversed by the Wnt inhibitor XAV-939. Single-cell analysis revealed that IL-33 overexpression skewed the immune microenvironment toward immunosuppression, while ST2 knockdown restored antitumor immunity. Our findings establish an IL-33-Wnt axis as a critical driver of breast cancer aggressiveness and propose AAV-mediated ST2 silencing as a novel therapeutic strategy. Targeting this axis may offer dual benefits by suppressing stemness and enhancing immune surveillance, warranting clinical exploration for advanced breast cancer.
Collapse
Affiliation(s)
- Guanglin Fan
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Shuting Zuo
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Zhen Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Siwei Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Liping Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Haoge Luo
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Yingdong Xie
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Yan Zhang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, People's Republic of China.
| | - Dong Li
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
2
|
Luo H, Liu L, Liu X, Xie Y, Huang X, Yang M, Shao C, Li D. Interleukin-33 (IL-33) promotes DNA damage-resistance in lung cancer. Cell Death Dis 2025; 16:274. [PMID: 40216748 PMCID: PMC11992107 DOI: 10.1038/s41419-025-07624-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025]
Abstract
Resistance to DNA damage is one of the primary mechanisms by which tumor cells evade the effects of standard chemotherapeutic agents and radiotherapy. Dynamic and complex interactions between the tumor microenvironment (TME) and tumor cells critically influence the DNA damage response. Interleukin-33 (IL-33) is a multifunctional cytokine secreted at high levels in response to cellular damage and stress. Recently, increasing evidence has suggested that IL-33 plays a key role in promoting the therapeutic resistance of tumors. However, the actual source of IL-33 during cancer therapy and how IL-33 contributes to a resistant TME remain incompletely understood. In this study, we found that both cancer-associated fibroblasts (CAFs) and tumor cells treated with DNA damage-inducing agents expressed and secreted high levels of IL-33, subsequently leading to enhanced DNA damage repair efficacy. Mechanistically, nuclear IL-33 primarily functions as a transcriptional co-activator of homologous recombination repair (HRR) genes, whereas the active form of IL-33 can drive the non-homologous end joining (NHEJ) pathway via the canonical IL-33/ST2 axis. Overall, we demonstrated that IL-33 plays a key role in mediating a DNA damage-resistant TME, which could represent a potential therapeutic vulnerability in chemoresistant cancer cells.
Collapse
Affiliation(s)
- Haoge Luo
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Liping Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xiaoping Liu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yingdong Xie
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xin Huang
- Department of Microbiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Ming Yang
- Department of Biochemistry & Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China
- Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Chen Shao
- Department of Biochemistry & Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, China.
- Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China.
| | - Dong Li
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China.
- Medical Basic Research Innovation Center of Airway Disease in North China, Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China.
| |
Collapse
|
3
|
Wang H, He K, Liu Y, Yang L, Wang Z, Wang H, Bai C, Liu J, Zhao L, Ma D, Liu Y. Expression and immune infiltration studies of IL-33-ST2-NF-κB signaling pathway in prostate cancer. Prostate 2024; 84:1398-1410. [PMID: 39113225 DOI: 10.1002/pros.24778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND To analyze the expression of interleukin-33 (IL-33), growth-stimulated expression gene 2 (ST2), nuclear factor-kappaB (NF-κB) and immune cell infiltration in prostate cancer, this study aims to provide an experimental basis for the clinical prevention and treatment of prostate cancer. METHODS The expression of IL-33 in PCa tissues was analyzed using TCGA, TIMER and HPA databases. Using the UALCAN database, the systematic exploration of the relationship between IL-33 and various clinicopathological parameters was conducted. The correlation between IL-33 expression and immune cell infiltration was investigated using TIMER, CIBERSORT and GEPIA databases. To verify these analyses, 22 cases of normal prostate (NP), 76 cases of benign prostatic hyperplasia (BPH), and 100 cases of PCa were recruited. Immunohistochemical staining was performed to examine the expression of IL-33, ST2, NF-κB, and the infiltration of immune cells. Correlations between these factors were then determined. RESULTS The expression of IL-33, ST2 and NF-κB was significantly lower in PCa tissues compared with NP (p < 0.05). IL-33 was not associated with age in PCa but showed associations with race, molecular characteristics, lymph node metastatic status, TP53 mutation and tumor grade. Furthermore, IL-33 was associated with immune cell infiltration. Positive correlations were observed between IL-33 and ST2 expressions, as well as between IL-33 and CD68+ macrophages in BPH and PCa. CONCLUSIONS IL-33, ST2 and NF-κB are lowly expressed in PCa tissues, their expression decreases with the increasing malignancy of cancer. IL-33, ST2 and NF-κB are factors associated with PCa immune infiltration. IL-33 has an inhibitory effect on prostate cancer through the IL-33/ST2/NF-κB signalling pathway.
Collapse
Affiliation(s)
- Han Wang
- School of Basic Medical College, Beihua University, Jilin, China
| | - Kang He
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Yuqi Liu
- School of Basic Medical College, Beihua University, Jilin, China
| | - Lijuan Yang
- School of Basic Medical College, Beihua University, Jilin, China
| | - Zhenjiang Wang
- School of Basic Medical College, Beihua University, Jilin, China
| | - Helin Wang
- School of Basic Medical College, Beihua University, Jilin, China
| | - Chengxia Bai
- School of Basic Medical College, Beihua University, Jilin, China
| | - Jian Liu
- People's hospital in Yushu city, Yushu, China
| | - Lijing Zhao
- Department of Rehabilitation, School of Nursing, Jilin University, Changchun, China
| | - Dongrui Ma
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yanbo Liu
- School of Basic Medical College, Beihua University, Jilin, China
| |
Collapse
|
4
|
Abdipourbozorgbaghi M, Vancura A, Radpour R, Haefliger S. Circulating miRNA panels as a novel non-invasive diagnostic, prognostic, and potential predictive biomarkers in non-small cell lung cancer (NSCLC). Br J Cancer 2024; 131:1350-1362. [PMID: 39215192 PMCID: PMC11473829 DOI: 10.1038/s41416-024-02831-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is characterised by its aggressiveness and poor prognosis. Early detection and accurate prediction of therapeutic responses remain critical for improving patient outcomes. In the present study, we investigated the potential of circulating microRNA (miRNA) as non-invasive biomarkers in patients with NSCLC. METHODS We quantified miRNA expression in plasma from 122 participants (78 NSCLC; 44 healthy controls). Bioinformatic tools were employed to identify miRNA panels for accurate NSCLC diagnosis. Validation was performed using an independent publicly available dataset of more than 4000 NSCLC patients. Next, we correlated miRNA expression with clinicopathological information to identify independent prognostic miRNAs and those predictive of anti-PD-1 treatment response. RESULTS We identified miRNA panels for lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC) diagnosis. The LUAD panel consists of seven circulating miRNAs (miR-9-3p, miR-96-5p, miR-147b-3p, miR-196a-5p, miR-708-3p, miR-708-5p, miR-4652-5p), while the LUSC panel comprises nine miRNAs (miR-130b-3p, miR-269-3p, miR-301a-5p, miR-301b-5p, miR-744-3p, miR-760, miR-767-5p, miR-4652-5p, miR-6499-3p). Additionally, miR-135b-5p, miR-196a-5p, miR-31-5p (LUAD), and miR-205 (LUSC) serve as independent prognostic markers for survival. Furthermore, two miRNA clusters, namely miR-183/96/182 and miR-767/105, exhibit predictive potential in anti-PD-1-treated LUAD patients. CONCLUSIONS Circulating miRNA signatures demonstrate diagnostic and prognostic value for NSCLC and may guide treatment decisions in clinical practice.
Collapse
Affiliation(s)
- Maryam Abdipourbozorgbaghi
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Adrienne Vancura
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Ramin Radpour
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Simon Haefliger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| |
Collapse
|
5
|
Radpour R, Simillion C, Wang B, Abbas HA, Riether C, Ochsenbein AF. IL-9 secreted by leukemia stem cells induces Th1-skewed CD4+ T cells, which promote their expansion. Blood 2024; 144:888-903. [PMID: 38941612 DOI: 10.1182/blood.2024024000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/30/2024] Open
Abstract
ABSTRACT In acute myeloid leukemia (AML), leukemia stem cells (LSCs) and leukemia progenitor cells (LPCs) interact with various cell types in the bone marrow (BM) microenvironment, regulating their expansion and differentiation. To study the interaction of CD4+ and CD8+ T cells in the BM with LSCs and LPCs, we analyzed their transcriptome and predicted cell-cell interactions by unbiased high-throughput correlation network analysis. We found that CD4+ T cells in the BM of patients with AML were activated and skewed toward T-helper (Th)1 polarization, whereas interleukin-9 (IL-9)-producing (Th9) CD4+ T cells were absent. In contrast to normal hematopoietic stem cells, LSCs produced IL-9, and the correlation modeling predicted IL9 in LSCs as a main hub gene that activates CD4+ T cells in AML. Functional validation revealed that IL-9 receptor signaling in CD4+ T cells leads to activation of the JAK-STAT pathway that induces the upregulation of KMT2A and KMT2C genes, resulting in methylation on histone H3 at lysine 4 to promote genome accessibility and transcriptional activation. This induced Th1-skewing, proliferation, and effector cytokine secretion, including interferon gamma (IFN-γ) and tumor necrosis factor α (TNF-α). IFN-γ and, to a lesser extent, TNF-α produced by activated CD4+ T cells induced the expansion of LSCs. In accordance with our findings, high IL9 expression in LSCs and high IL9R, TNF, and IFNG expression in BM-infiltrating CD4+ T cells correlated with worse overall survival in AML. Thus, IL-9 secreted by AML LSCs shapes a Th1-skewed immune environment that promotes their expansion by secreting IFN-γ and TNF-α.
Collapse
MESH Headings
- Interleukin-9/genetics
- Interleukin-9/metabolism
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/immunology
- Th1 Cells/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Tumor Microenvironment/immunology
- Receptors, Interleukin-9/genetics
- Receptors, Interleukin-9/metabolism
- Interferon-gamma/metabolism
- Histone-Lysine N-Methyltransferase/genetics
Collapse
Affiliation(s)
- Ramin Radpour
- Department for BioMedical Research, Tumor Immunology, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Bofei Wang
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
| | - Hussein A Abbas
- Department of Leukemia, MD Anderson Cancer Center, Houston, TX
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX
| | - Carsten Riether
- Department for BioMedical Research, Tumor Immunology, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Adrian F Ochsenbein
- Department for BioMedical Research, Tumor Immunology, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
6
|
Pathania AS. Immune Microenvironment in Childhood Cancers: Characteristics and Therapeutic Challenges. Cancers (Basel) 2024; 16:2201. [PMID: 38927907 PMCID: PMC11201451 DOI: 10.3390/cancers16122201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/23/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The tumor immune microenvironment is pivotal in cancer initiation, advancement, and regulation. Its molecular and cellular composition is critical throughout the disease, as it can influence the balance between suppressive and cytotoxic immune responses within the tumor's vicinity. Studies on the tumor immune microenvironment have enriched our understanding of the intricate interplay between tumors and their immunological surroundings in various human cancers. These studies illuminate the role of significant components of the immune microenvironment, which have not been extensively explored in pediatric tumors before and may influence the responsiveness or resistance to therapeutic agents. Our deepening understanding of the pediatric tumor immune microenvironment is helping to overcome challenges related to the effectiveness of existing therapeutic strategies, including immunotherapies. Although in the early stages, targeted therapies that modulate the tumor immune microenvironment of pediatric solid tumors hold promise for improved outcomes. Focusing on various aspects of tumor immune biology in pediatric patients presents a therapeutic opportunity that could improve treatment outcomes. This review offers a comprehensive examination of recent literature concerning profiling the immune microenvironment in various pediatric tumors. It seeks to condense research findings on characterizing the immune microenvironment in pediatric tumors and its impact on tumor development, metastasis, and response to therapeutic modalities. It covers the immune microenvironment's role in tumor development, interactions with tumor cells, and its impact on the tumor's response to immunotherapy. The review also discusses challenges targeting the immune microenvironment for pediatric cancer therapies.
Collapse
Affiliation(s)
- Anup Singh Pathania
- Department of Biochemistry and Molecular Biology, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
7
|
Wei L, He P, Tan Z, Lin C, Wei Z. Comprehensively analysis of IL33 in hepatocellular carcinoma prognosis, immune microenvironment and biological role. J Cell Mol Med 2024; 28:e18468. [PMID: 38923705 PMCID: PMC11196832 DOI: 10.1111/jcmm.18468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/22/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
IL33 plays an important role in cancer. However, the role of liver cancer remains unclear. Open-accessed data was obtained from the Cancer Genome Atlas, Xena, and TISCH databases. Different algorithms and R packages are used to perform various analyses. Here, in our comprehensive study on IL33 in HCC, we observed its differential expression across cancers, implicating its role in cancer development. The single-cell analysis highlighted its primary expression in endothelial cells, unveiling correlations within the HCC microenvironment. Also, the expression level of IL33 was correlated with patients survival, emphasizing its potential prognostic value. Biological enrichment analyses revealed associations with stem cell division, angiogenesis, and inflammatory response. IL33's impact on the immune microenvironment showcased correlations with diverse immune cells. Genomic features and drug sensitivity analyses provided insights into IL33's broader implications. In a pan-cancer context, IL33 emerged as a potential tumour-inhibitor, influencing immune-related molecules. This study significantly advances our understanding of IL33 in cancer biology. IL33 exhibited differential expression across cancers, particularly in endothelial cells within the HCC microenvironment. IL33 is correlated with the survival of HCC patients, indicating potential prognostic value and highlighting its broader implications in cancer biology.
Collapse
Affiliation(s)
- Lifang Wei
- Health Management CenterThe Affiliated Hospital of Youjiang Medical University for NationalitiesGuangxiChina
| | - Ping He
- School of Laboratory MedicineYoujiang Medical University for NationalitiesGuangxiChina
| | - Zhongqiu Tan
- Department of OncologyThe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - Cheng Lin
- Department of OncologyThe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
| | - Zhongheng Wei
- Department of OncologyThe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseGuangxiChina
- Guangxi Clinical Medical Research Center for Hepatobiliary DiseasesThe Affiliated Hospital of Youjiang Medical University for NationalitiesBaiseChina
| |
Collapse
|
8
|
Karsten H, Matrisch L, Cichutek S, Fiedler W, Alsdorf W, Block A. Broadening the horizon: potential applications of CAR-T cells beyond current indications. Front Immunol 2023; 14:1285406. [PMID: 38090582 PMCID: PMC10711079 DOI: 10.3389/fimmu.2023.1285406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Engineering immune cells to treat hematological malignancies has been a major focus of research since the first resounding successes of CAR-T-cell therapies in B-ALL. Several diseases can now be treated in highly therapy-refractory or relapsed conditions. Currently, a number of CD19- or BCMA-specific CAR-T-cell therapies are approved for acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), multiple myeloma (MM), and follicular lymphoma (FL). The implementation of these therapies has significantly improved patient outcome and survival even in cases with previously very poor prognosis. In this comprehensive review, we present the current state of research, recent innovations, and the applications of CAR-T-cell therapy in a selected group of hematologic malignancies. We focus on B- and T-cell malignancies, including the entities of cutaneous and peripheral T-cell lymphoma (T-ALL, PTCL, CTCL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), chronic lymphocytic leukemia (CLL), classical Hodgkin-Lymphoma (HL), Burkitt-Lymphoma (BL), hairy cell leukemia (HCL), and Waldenström's macroglobulinemia (WM). While these diseases are highly heterogenous, we highlight several similarly used approaches (combination with established therapeutics, target depletion on healthy cells), targets used in multiple diseases (CD30, CD38, TRBC1/2), and unique features that require individualized approaches. Furthermore, we focus on current limitations of CAR-T-cell therapy in individual diseases and entities such as immunocompromising tumor microenvironment (TME), risk of on-target-off-tumor effects, and differences in the occurrence of adverse events. Finally, we present an outlook into novel innovations in CAR-T-cell engineering like the use of artificial intelligence and the future role of CAR-T cells in therapy regimens in everyday clinical practice.
Collapse
Affiliation(s)
- Hendrik Karsten
- Faculty of Medicine, University of Hamburg, Hamburg, Germany
| | - Ludwig Matrisch
- Department of Rheumatology and Clinical Immunology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Faculty of Medicine, University of Lübeck, Lübeck, Germany
| | - Sophia Cichutek
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Winfried Alsdorf
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Andreas Block
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| |
Collapse
|