1
|
Caloian AD, Cristian M, Calin E, Pricop AR, Mociu SI, Seicaru L, Deacu S, Ciufu N, Suceveanu AI, Suceveanu AP, Mazilu L. Epigenetic Symphony in Diffuse Large B-Cell Lymphoma: Orchestrating the Tumor Microenvironment. Biomedicines 2025; 13:853. [PMID: 40299416 PMCID: PMC12024808 DOI: 10.3390/biomedicines13040853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Abstract
DLBCL is a testament to the complexity of nature. It is characterized by remarkable diversity in its molecular and pathological subtypes and clinical manifestations. Despite the strides made in DLBCL treatment and the introduction of innovative drugs, around one-third of patients face a relapse or develop refractory disease. Recent findings over the past ten years have highlighted the critical interplay between the evolution of DLBCL and various epigenetic mechanisms, including chromatin remodeling, DNA methylation, histone modifications, and the regulatory roles of non-coding RNAs. These epigenetic alterations are integral to the pathways of oncogenesis, tumor progression, and the development of therapeutic resistance. In the past decade, the identification of dysregulated epigenetic mechanisms in lymphomas has paved the way for an exciting field of epigenetic therapies. Crucially, these epigenetic transformations span beyond tumor cells to include the sophisticated network within the tumor microenvironment (TME). While the exploration of epigenetic dysregulation in lymphoma cells is thriving, the mechanisms affecting the functions of immune cells in the TME invite further investigation. This review is dedicated to weaving together the narrative of epigenetic alterations impacting both lymphoma cells with a focus on their infiltrating immune companions.
Collapse
Affiliation(s)
- Andreea-Daniela Caloian
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Hemato-Oncology, “Ovidius” Clinical Hospital, 900470 Constanta, Romania;
| | - Miruna Cristian
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology-CEDMOG, “Ovidius” University of Constanta, 900470 Constanta, Romania
- Department of Forensic Medicine, “Sf. Apostol Andrei” Emergency County Hospital, 900439 Constanta, Romania
| | - Elena Calin
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Hemato-Oncology, “Ovidius” Clinical Hospital, 900470 Constanta, Romania;
| | - Andreea-Raluca Pricop
- Department of Dermatology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania;
| | - Stelian-Ilie Mociu
- Department of Hemato-Oncology, “Ovidius” Clinical Hospital, 900470 Constanta, Romania;
| | - Liliana Seicaru
- Department of Clinical Patology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania;
| | - Sorin Deacu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Clinical Patology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania;
| | - Nicolae Ciufu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Hemato-Oncology, “Ovidius” Clinical Hospital, 900470 Constanta, Romania;
| | - Andra-Iulia Suceveanu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Gastroenterology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
| | - Adrian-Paul Suceveanu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Gastroenterology, “Sf. Apostol Andrei” Emergency County Hospital, 900591 Constanta, Romania
| | - Laura Mazilu
- Faculty of Medicine, “Ovidius” University of Constanta, 900470 Constanta, Romania; (E.C.); (S.D.); (N.C.); (A.-I.S.); (A.-P.S.); (L.M.)
- Department of Hemato-Oncology, “Ovidius” Clinical Hospital, 900470 Constanta, Romania;
| |
Collapse
|
2
|
Chen P, Wang H, Tang Z, Shi J, Cheng L, Zhao C, Li X, Zhou C. Selective Depletion of CCR8+Treg Cells Enhances the Antitumor Immunity of Cytotoxic T Cells in Lung Cancer by Dendritic Cells. J Thorac Oncol 2025:S1556-0864(25)00109-1. [PMID: 40056978 DOI: 10.1016/j.jtho.2025.02.029] [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: 09/24/2024] [Revised: 01/17/2025] [Accepted: 02/22/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION Accumulation of regulatory T (Treg) cells, an immunosuppressive population, limits the efficacy of immunotherapy in NSCLC. C-C motif chemokine receptor 8 (CCR8) is selectively expressed in tumor-infiltrating Treg cells and is, therefore, considered an ideal target. METHODS The efficacy and safety of anti-CCR8 monotherapy and its combination with programmed cell death protein-1 (PD1) inhibitor were evaluated in four NSCLC-bearing mice models. To track the dynamic changes in tumor microenvironment, we performed the single-cell RNA sequencing, the single-cell T-cell receptor sequencing analysis, the flow cytometry, the multi-color immunofluorescence, and the Luminex assay on tumors after three, seven, 14, and 21 days of different treatment regimens. Then, in vitro and in vivo experiments were applied to validate our findings and explore molecular mechanisms of the synergistic effects. RESULTS Across four NSCLC-bearing mice models, the combination of CCR8 antibody and PD1 inhibitor significantly reduced tumor growth (p < 0.05) without obvious mouse body weight drops and systemic cytokine storm. The anti-CCR8 therapy synergizes with PD1 blockade by remodeling the tumor microenvironment and disrupting CCR8+Treg-C-C motif chemokine ligand 5 (CCL5)+ dendritic cells (DC) interaction. Mechanistically, therapeutic depletion of CCR8+Treg cells combined with PD1 inhibitor extremely increased interleukin-12 secretion by the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway activation on CCL5+ DCs, thereby promoting cytotoxic activity of CD8+ T cells. The therapeutic potential of the CCR8 antibody LM-108 in combination with immunotherapy was observed in clinical patients with advanced NSCLC. CONCLUSION Overall, CCR8 expression on tumor-infiltrating Treg cells is correlated with immunosuppressive function on DCs and CD8+ T cells, thus impeding antitumor immunity.
Collapse
Affiliation(s)
- Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Haowei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Zhuoran Tang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Jinpeng Shi
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Lei Cheng
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Chao Zhao
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China; Department of Medical Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China.
| |
Collapse
|
3
|
Cui Y, Hu G, Han X, Li W, Wang X, Qian Z, Li L, Qiu L, Zhou S, Zhang H. Comprehensive Analysis of Single-Cell and Bulk Transcriptomics Identified Regulatory T-Cell Features as Predictors of Prognosis in Diffuse Large B-Cell Lymphoma. Hematol Oncol 2025; 43:e70050. [PMID: 40041930 DOI: 10.1002/hon.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 05/12/2025]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a biologically and clinically heterogeneous malignancy. Advances in transcriptomic and genetic profiling have significantly enhanced our understanding of the disease's intrinsic pathogenesis, uncovering numerous potential therapeutic targets. However, the impact of tumor-infiltrating Regulatory T cells (Tregs) on the prognosis of DLBCL remains controversial. Here, we developed a Treg-associated gene signature by integrating single-cell and bulk transcriptome data to predict the prognosis of DLBCL patients receiving standard immunochemotherapy. In total, 227 Tregs feature genes were identified, six of which were selected for constructing a prognostic signature. DLBCL patients possessing high-risk scores had significantly poorer survival outcomes than those who possess low-risk scores in NCICCR and validation cohorts. Mutations in PIM1, MYD88, DTX1, CARD11, CD79B, ETV6, BCL6, and CDKN2A were predominantly observed in the high-risk group, whereas alterations in TNFRSF14 and DNMT3A were more frequently detected in the low-risk group. Immune infiltration analysis revealed that the high-risk group exhibited an immunosuppressive microenvironment, whereas the low-risk group showed a higher abundance of non-cellular components in the tumor microenvironment (TME). Finally, the Treg features TNFRSF25 and SELL can effectively predict long-term responses to Axicabtagene Ciloleucel (Axi-cel) treatment. In summary, our study developed a prognostic signature consisting of six Treg feature genes by integrating single-cell and bulk transcriptomics to predict clinical outcomes in DLBCL patients. The risk signature was significantly associated with immunological characteristics.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
- Prognosis
- Transcriptome
- Female
- Male
- Gene Expression Profiling
- Middle Aged
- Single-Cell Analysis/methods
- Tumor Microenvironment
- Biomarkers, Tumor/genetics
- Aged
- Adult
Collapse
Affiliation(s)
- Yaoli Cui
- Department of Lymphoma and National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Ge Hu
- Department of Lymphoma and National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Xue Han
- Department of Lymphoma and National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Wei Li
- Department of Lymphoma and National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Xianhuo Wang
- Department of Lymphoma and National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Zhengzi Qian
- Department of Lymphoma and National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Lanfang Li
- Department of Lymphoma and National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Lihua Qiu
- Department of Lymphoma and National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Shiyong Zhou
- Department of Lymphoma and National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| | - Huilai Zhang
- Department of Lymphoma and National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, the Sino-US Center for Lymphoma and Leukemia Research, Tianjin, China
| |
Collapse
|
4
|
Li S, Pan Y, Ye R, Wang Y, Li L. Immune checkpoints in B-cell Lymphoma: Still an Unmet challenge from Basic research to clinical practice. Int Immunopharmacol 2025; 146:113717. [PMID: 39673995 DOI: 10.1016/j.intimp.2024.113717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 12/16/2024]
Abstract
In the last decade, advancements in immunotherapy knowledge have highlighted CTLA-4, PD-1, LAG-3, TIM-3, and TIGIT, decisive immune checkpoints exhibiting within the tumor microenvironment (TME), as fundamental objects for cancer immunotherapy. The widespread clinical use of immune checkpoint inhibitors (ICls), employing PD-1/PD-L1 or CTLA-4 antibodies to obstruct crucial checkpoint regulators, is noted in treating B-cell lymphoma patients. Nevertheless, the prolonged advantages of the currently employed treatments against CTLA-4, PD-1, and PD-L1 are uncommon among patients. Thus, recent focus has been progressively moved to additional immune checkpoints on T cells, like LAG-3, TIM-3, and TIGIT, which are now seen as reassuring targets for treatment and broadly acknowledged. There are several types of immunecheckpoint molecules expressed by T cells, and inhibitors targeting immune checkpoints can revive and amplify the immune response of T lymphocytes against tumors, a crucial aspect in lymphoma therapy. However, there is little knowledge about their regulation. Herein, we discuss the anti-tumor effects and functions of ICIs in controlling T-cell activity, as well as the progress in combined application with other immunotherapies.
Collapse
Affiliation(s)
- Sijia Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Yuanyuan Pan
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Ruyu Ye
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Yu Wang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Li Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China.
| |
Collapse
|
5
|
Sun W, Hu S, Wang X. Advances and clinical applications of immune checkpoint inhibitors in hematological malignancies. Cancer Commun (Lond) 2024; 44:1071-1097. [PMID: 39073258 PMCID: PMC11492363 DOI: 10.1002/cac2.12587] [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: 11/30/2023] [Revised: 06/09/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Immune checkpoints are differentially expressed on various immune cells to regulate immune responses in tumor microenvironment. Tumor cells can activate the immune checkpoint pathway to establish an immunosuppressive tumor microenvironment and inhibit the anti-tumor immune response, which may lead to tumor progression by evading immune surveillance. Interrupting co-inhibitory signaling pathways with immune checkpoint inhibitors (ICIs) could reinvigorate the anti-tumor immune response and promote immune-mediated eradication of tumor cells. As a milestone in tumor treatment, ICIs have been firstly used in solid tumors and subsequently expanded to hematological malignancies, which are in their infancy. Currently, immune checkpoints have been investigated as promising biomarkers and therapeutic targets in hematological malignancies, and novel immune checkpoints, such as signal regulatory protein α (SIRPα) and tumor necrosis factor-alpha-inducible protein 8-like 2 (TIPE2), are constantly being discovered. Numerous ICIs have received clinical approval for clinical application in the treatment of hematological malignancies, especially when used in combination with other strategies, including oncolytic viruses (OVs), neoantigen vaccines, bispecific antibodies (bsAb), bio-nanomaterials, tumor vaccines, and cytokine-induced killer (CIK) cells. Moreover, the proportion of individuals with hematological malignancies benefiting from ICIs remains lower than expected due to multiple mechanisms of drug resistance and immune-related adverse events (irAEs). Close monitoring and appropriate intervention are needed to mitigate irAEs while using ICIs. This review provided a comprehensive overview of immune checkpoints on different immune cells, the latest advances of ICIs and highlighted the clinical applications of immune checkpoints in hematological malignancies, including biomarkers, targets, combination of ICIs with other therapies, mechanisms of resistance to ICIs, and irAEs, which can provide novel insight into the future exploration of ICIs in tumor treatment.
Collapse
Affiliation(s)
- Wenyue Sun
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
| | - Shunfeng Hu
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
| | - Xin Wang
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
- Taishan Scholars Program of Shandong ProvinceJinanShandongP. R. China
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongP. R. China
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuP. R. China
| |
Collapse
|
6
|
Meyiah A, Elkord E. What is the relevance of FoxP3 in the tumor microenvironment and cancer outcomes? Expert Rev Clin Immunol 2024; 20:803-809. [PMID: 38512803 DOI: 10.1080/1744666x.2024.2334258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/20/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION Forkhead box P3 (FoxP3) transcription factor plays critical roles in controlling immune responses and cancer progression in different cancers. FoxP3 expression within the tumor microenvironment (TME) may influence clinical outcomes negatively or positively, and it could play dual roles in cancer, either by promoting or inhibiting tumor development and progression. Some studies reported that high levels of FoxP3 could be associated with tumor progression and worse prognosis, while others reported contradictory results. AREAS COVERED In this special report, we present a brief account on the role and function of FoxP3 in the TME, and its contribution to the clinical outcomes of cancer patients. Importantly, we give insights on the potential factors that could contribute to different clinical outcomes in cancer patients. EXPERT OPINION Different studies showed that FoxP3 expression can be associated with bad prognoses in cancer patients. However, FoxP3 could have opposing roles by enhancing cancer progression or regression. Location and expression of FoxP3 in T cells or tumor cells can have different impacts on cancer prognoses. Different factors should be considered to establish FoxP3 as a more robust prognostic biomarker and a potential therapeutic target for enhancing anti-tumor immunity and improving clinical outcomes of cancer patients.
Collapse
Affiliation(s)
- Abdo Meyiah
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Eyad Elkord
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, UK
| |
Collapse
|
7
|
Koumpis E, Papoudou-Bai A, Papathanasiou K, Kolettas E, Kanavaros P, Hatzimichael E. Unraveling the Immune Microenvironment in Diffuse Large B-Cell Lymphoma: Prognostic and Potential Therapeutic Implications. Curr Issues Mol Biol 2024; 46:7048-7064. [PMID: 39057061 PMCID: PMC11276293 DOI: 10.3390/cimb46070420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Diffuse large B cell lymphoma (DLBCL) is a multifaceted condition characterized by significant diversity in its molecular and pathological subtypes and clinical manifestation. Despite the progress made in the treatment of DLBCL through the development of novel drugs, an estimated one-third of patients encounter relapse or acquire refractory disease. The tumor microenvironment (TME) of DLBCL, a complex network consisting of cellular and noncellular components that engage in interactions with the tumor, is a parameter that is gaining increasing attention. The TME comprises both the immune and nonimmune microenvironments. The immune microenvironment comprises natural killer (NK) cells, dendritic cells (DCs), tumor-associated macrophages (TAMs), neutrophils, myeloid-derived suppressor cells (MDSCs), and T and B lymphocytes. The nonimmune microenvironment consists of the extracellular matrix (ECM), cancer-associated fibroblasts (CAFs), mesenchymal stromal cells, and other molecules that are secreted. Despite ongoing research, the exact impact of these components and their interaction on the progression of the disease remains elusive. A comprehensive review of significant discoveries concerning the cellular and noncellular constituents, molecular characteristics, and treatment response and prognosis of the TME in DLBCL, as well as the potential targeting of the TME with novel therapeutic approaches, is provided in this article.
Collapse
Affiliation(s)
- Epameinondas Koumpis
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 500 Ioannina, Greece; (E.K.); (K.P.)
| | - Alexandra Papoudou-Bai
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 500 Ioannina, Greece;
| | - Konstantina Papathanasiou
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 500 Ioannina, Greece; (E.K.); (K.P.)
| | - Evangelos Kolettas
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 110 Ioannina, Greece;
- Biomedical Research Institute, Foundation for Research and Technology, 45 110 Ioannina, Greece
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 110 Ioannina, Greece;
| | - Eleftheria Hatzimichael
- Department of Hematology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45 500 Ioannina, Greece; (E.K.); (K.P.)
| |
Collapse
|
8
|
Bai X, Lu F, Li S, Zhao Z, Wang N, Zhao Y, Ma G, Zhang F, Su X, Wang D, Ye J, Li P, Ji C. Cuproptosis-related lncRNA signature as a prognostic tool and therapeutic target in diffuse large B cell lymphoma. Sci Rep 2024; 14:12926. [PMID: 38839842 PMCID: PMC11153514 DOI: 10.1038/s41598-024-63433-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Cuproptosis is a newly defined form of programmed cell death that relies on mitochondria respiration. Long noncoding RNAs (lncRNAs) play crucial roles in tumorigenesis and metastasis. However, whether cuproptosis-related lncRNAs are involved in the pathogenesis of diffuse large B cell lymphoma (DLBCL) remains unclear. This study aimed to identify the prognostic signatures of cuproptosis-related lncRNAs in DLBCL and investigate their potential molecular functions. RNA-Seq data and clinical information for DLBCL were collected from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Cuproptosis-related lncRNAs were screened out through Pearson correlation analysis. Utilizing univariate Cox, least absolute shrinkage and selection operator (Lasso) and multivariate Cox regression analysis, we identified seven cuproptosis-related lncRNAs and developed a risk prediction model to evaluate its prognostic value across multiple groups. GO and KEGG functional analyses, single-sample GSEA (ssGSEA), and the ESTIMATE algorithm were used to analyze the mechanisms and immune status between the different risk groups. Additionally, drug sensitivity analysis identified drugs with potential efficacy in DLBCL. Finally, the protein-protein interaction (PPI) network were constructed based on the weighted gene co-expression network analysis (WGCNA). We identified a set of seven cuproptosis-related lncRNAs including LINC00294, RNF139-AS1, LINC00654, WWC2-AS2, LINC00661, LINC01165 and LINC01398, based on which we constructed a risk model for DLBCL. The high-risk group was associated with shorter survival time than the low-risk group, and the signature-based risk score demonstrated superior prognostic ability for DLBCL patients compared to traditional clinical features. By analyzing the immune landscapes between two groups, we found that immunosuppressive cell types were significantly increased in high-risk DLBCL group. Moreover, functional enrichment analysis highlighted the association of differentially expressed genes with metabolic, inflammatory and immune-related pathways in DLBCL patients. We also found that the high-risk group showed more sensitivity to vinorelbine and pyrimethamine. A cuproptosis-related lncRNA signature was established to predict the prognosis and provide insights into potential therapeutic strategies for DLBCL patients.
Collapse
Affiliation(s)
- Xiaoran Bai
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Department of Lymphoma and Plasmacytoma Disease, Senior Department of Hematology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Fei Lu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Shuying Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Zhe Zhao
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Nana Wang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yanan Zhao
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Guangxin Ma
- Hematology and Oncology Unit, Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Fan Zhang
- Gastroenterology Intensive Care Unit, Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xiuhua Su
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Dongmei Wang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Jingjing Ye
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Peng Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Chunyan Ji
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| |
Collapse
|
9
|
Zheng S, Ma J, Li J, Pang X, Ma M, Ma Z, Cui W. Lower PTEN may be associated with CD8+ T cell exhaustion in diffuse large B-cell lymphoma. Hum Immunol 2023; 84:551-560. [PMID: 37481380 DOI: 10.1016/j.humimm.2023.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/30/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Initially discovered in chronic viral infection and then extended to tumor, 'T-cell exhaustion' is a broad term describing the response of T cells to chronic antigen stimulation. By definition, whether T-cell exhaustion occurs in diffuse large B-cell lymphoma (DLBCL) remains largely unknown because little has been described. Here, the immune-suppressing checkpoint molecules involved in T-cell exhaustion, including PD-1, PD-L1, TIM-3 and TIGIT, whose expression levels were analyzed in DLBCL, were retrieved from the GEPIA database. Compared with the normal control, CD8A, TNFA, IFNG and GZMA were markedly elevated in DLBCL, indicating that infiltrated CD8+ T cells predominate in DLBCL. Meanwhile, inhibitory immune checkpoints, such as PD-1, PD-L1, TIGIT and TIM-3 were drastically higher in DLBCL. PTEN, WNT2 and DKK3 expression were also appraised. It was revealed that PTEN was lower in DLBCL, without being statistically significant. In contrast with PTEN, DKK3 and WNT2 were shown to be pronouncedly higher in DLBCL relative to the normal control. Prognostically, only TIGIT was found to be associated with overall survival in DLBCL. Collectively, all the data we curetted from the GEPIA and TIMER 2.0 databases explicitly indicate that CD8+ T cell exhaustion took place, which may be linked with lower PTEN in DLBCL. To the best of our knowledge, this is the first bioinformatic report explicitly proposing that CD8+ T cell exhaustion occurs in DLBCL, which may be associated with lower PTEN.
Collapse
Affiliation(s)
- Shutao Zheng
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Jiajia Ma
- The Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, PR China
| | - Junna Li
- The Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, PR China
| | - Xuelian Pang
- The Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, PR China
| | - Mingfu Ma
- The Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, PR China
| | - Zhiping Ma
- The Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, PR China
| | - Wenli Cui
- The Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, 830011 Urumqi, Xinjiang, PR China.
| |
Collapse
|
10
|
Stirm K, Leary P, Wüst D, Stark D, Joller N, Karakus U, Boyman O, Tzankov A, Müller A. Treg-selective IL-2 starvation synergizes with CD40 activation to sustain durable responses in lymphoma models. J Immunother Cancer 2023; 11:e006263. [PMID: 36822670 PMCID: PMC9950978 DOI: 10.1136/jitc-2022-006263] [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] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Roughly half of all diffuse large B-cell lymphomas (DLBCLs) are infiltrated by large numbers of regulatory T-cells (Tregs). Although the presence of 'effector' Tregs in particular is associated with an inferior prognosis in patients on standard rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) immunochemotherapy, the role of this cell type during lymphoma initiation and progression is poorly understood. METHODS Here, we use tissue microarrays containing prospectively collected DLBCL patient specimens, as well as data from publicly available cohorts to explore the mutational landscape of Treg-infiltrated DLBCL. We further take advantage of a model of MYC-driven lymphoma to mechanistically dissect the contribution of Tregs to lymphoma pathogenesis and to develop a strategy of Treg-selective interleukin-2 (IL-2) starvation to improve immune control of MYC-driven lymphoma. RESULTS We find that all genetic DLBCL subtypes, except for one characterized by co-occurring MYD88/CD79 mutations, are heavily infiltrated by Tregs. Spectral flow cytometry and scRNA-sequencing reveal the robust expression of functional and immunosuppressive markers on Tregs infiltrating MYC-driven lymphomas; notably, we find that intratumoral Tregs arise due to local conversion from naïve CD4+ precursors on tumor contact. Treg ablation in Foxp3iDTR mice, or by antibody-mediated Treg-selective blockade of IL-2 signaling, strongly reduces the lymphoma burden. We identify lymphoma B-cells as a major source of IL-2, and show that the effects of Treg depletion are reversed by the simultaneous depletion of Foxp3-negative CD4+ T-cells, but not CD8+ T-cells or natural killer (NK) cells. The inhibition of ATP hydrolyzation and adenosine production by Tregs at least partly phenocopies the effects of Treg depletion. Treg depletion further synergizes with pro-apoptotic CD40 activation to sustain durable responses. CONCLUSION The combined data implicate Tregs as a potential therapeutic target in DLBCL, especially in combination with other immunotherapies.
Collapse
Affiliation(s)
- Kristin Stirm
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Peter Leary
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Daria Wüst
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Dominique Stark
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Nicole Joller
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Ufuk Karakus
- Department of Immunology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Alexandar Tzankov
- Institute of Medical Genetics and Pathology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Ma J, Pang X, Li J, Zhang W, Cui W. The immune checkpoint expression in the tumor immune microenvironment of DLBCL: Clinicopathologic features and prognosis. Front Oncol 2022; 12:1069378. [PMID: 36561512 PMCID: PMC9763555 DOI: 10.3389/fonc.2022.1069378] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
Background & aims The immune checkpoint recently provides a new strategy for the immunotherapy of malignant tumors. However, the role in the immune microenvironment of DLBCL is not completely clear. Methods We detected the expression of PD-1, LAG-3, TIM-3, and TIGIT on TILs and on tumor cells among 174 DLBCL patients by IHC. Results In TILs, the positive rates of PD-1, LAG-3, TIM-3 and TIGIT were 79.3%, 78.8%, 62.7% and 69.5%, respectively.TIM-3 and TIGIT were expressed in 44.8% and 45.4% of tumor cells. The expression of TIM-3 in TILs was significantly correlated with the Ann-Arbor stage (P=0.039). There was a positive correlation Between PD-1 and LAG-3 or TIM-3 and TIGIT.In addition, LAG-3 expression in TILs was associated with inferior prognosis.Multivariate analysis showed that PS score and R-CHOP therapy were independent risk factors for OS and PFS in patients with DLBCL (P=0.000). Conclusions The expression level of TIM-3 is closely related to the Ann-Arbor stage, which may be expected to be a new index to evaluate the invasiveness of DLBCL. PD-1 was correlated with the expression of LAG-3, and the high expression of LAG-3 and LAG-3/PD-1 predicted the poor prognosis of DLBCL. Therefore, LAG-3 may become a new target of immunotherapy, or be used in combination with PD-1 inhibitors to improve the drug resistance of current patients with DLBCL.
Collapse
|
12
|
Wu H, Sun HC, Ouyang GF. T-cell immunoglobulin mucin molecule-3, transformation growth factor β, and chemokine-12 and the prognostic status of diffuse large B-cell lymphoma. World J Clin Cases 2022; 10:11804-11811. [PMID: 36405294 PMCID: PMC9669877 DOI: 10.12998/wjcc.v10.i32.11804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/08/2022] [Accepted: 10/19/2022] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The effects of T-cell immunoglobulin mucin molecule-3 (Tim-3), transforming growth factor β (TGF-β), and chemokine-12 (CXCL12) expression on the prognosis of patients with diffuse large B-cell lymphoma (DLBCL) have not been elucidated.
AIM To examine the correlation between Tim-3, TGF-β and CXCL12 expression and DLBCL prognosis.
METHODS Lymph node tissues of 97 patients with DLBCL and 93 normal-response hyperplastic lymph node tissues treated from January 2017 to May 2019 were selected as the DLBCL and control groups, respectively. The expression of Tim-3, TGF-β, and CXCL12 was detected immunohistochemically. Patients were followed up for 3 years, and progression-free survival was recorded. Cox multifactorial analysis was performed to analyze the risk factors for poor prognosis.
RESULTS The positive expression rates of Tim-3, TGF-β, and CXCL12 were higher in DLBCL tissues than in non-cancerous (control) tissues (P < 0.05). One-year post-surgery, the positive expression rates of Tim-3, TGF-β, and CXCL12 were higher in patients with effective treatment than in those with ineffective treatment (P < 0.05). The 3-year progression-free survival of 97 patients with DLBCL was 67.01% (65/97). Univariate analysis revealed that clinical stage, bone marrow infiltration, International Prognostic Index (IPI) score, Tim-3 positivity, TGF-β positivity, and CXCL12 positivity were associated with poor prognosis (P < 0.05). Multivariate Cox regression analysis demonstrated that clinical stage III–IV, bone marrow infiltration, mediate-to-high-risk IPI scores, Tim-3 positivity, TGF-β positivity, and CXCL12 positivity were independent risk factors affecting prognosis (P < 0.05).
CONCLUSION DLBCL tissues exhibit high positive expression of Tim-3, TGF-β, and CXCL12, and a high expression of all three indicates a poor prognosis.
Collapse
Affiliation(s)
- Hao Wu
- Department of Hematology, Ningbo First Hospital, Ningbo Clinical Research Center for Hematologic Malignancies, Ningbo 315010, Zhejiang Province, China
| | - Hui-Cong Sun
- Department of Adult Internal Medicine, Ningbo Women and Children's Hospital, Ningbo 315012, Zhejiang Province, China`
| | - Gui-Fang Ouyang
- Department of Hematology, Ningbo First Hospital, Ningbo Clinical Research Center for Hematologic Malignancies, Ningbo 315010, Zhejiang Province, China
| |
Collapse
|
13
|
Qiu J, Xu L, Zeng X, Wu H, Liang F, Lv Q, Du Z. CCL5 mediates breast cancer metastasis and prognosis through CCR5/Treg cells. Front Oncol 2022; 12:972383. [PMID: 36033472 PMCID: PMC9399502 DOI: 10.3389/fonc.2022.972383] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022] Open
Abstract
Background and aims CCL5 is considered to contribute to the biological function of a variety of cancer types, but its specific mechanism is still unclear. This study aimed to reveal the mechanism of CCL5 in the invasion, metastasis, and prognosis of breast cancer. Methods The expression of CCL5 in tumor tissue and serum was measured with a Luminex protein detection kit, and the correlation between CCL5 and clinical parameters was evaluated. Kaplan–Meier analysis was used to analyze the effect of CCL5 on the prognosis of breast cancer patients. Protein interaction network analysis and gene coexpression were used to determine the receptor that has the strongest interaction with CCL5. Enrichment analysis was used to study the possible pathway by which CCL5 affects breast cancer progression. We used immunofluorescence staining and flow cytometry to estimate the fraction of immunity-related components in the tumor microenvironment. Results The expression level of CCL5 in breast cancer patients was positively correlated with the degree of axillary lymph node metastasis; CCL5 in tumor tissue was correlated with estrogen receptor status (P = 0.034), progesterone receptor (P = 0.009), nuclear grade (P = 0.013), clinical stage (P < 0.001) and molecular subtype (P = 0.024) in breast cancer patients. Breast cancer patients with high CCL5 expression had worse disease-free survival (P = 0.031) and breast cancer-specific survival (P = 0.043); however, CCL5 had no effect on overall survival (P = 0.077). CCL5 affected tumor progression through CCR5, and the T-cell-related immune pathway may be the main pathway; the CD4+/CD8+, CCR5+/CD4+ and Treg/CCR5+ cell ratios were significantly increased in the lymph node metastasis group. Conclusion CCL5 affects the Treg/CD4+CCR5+ cell ratio in breast cancer patients through CCR5, thus affecting breast cancer metastasis and prognosis.
Collapse
Affiliation(s)
- Juanjuan Qiu
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Transplant Engineering and Immunology NHC, West China Hospital, Sichuan University, Chengdu, China
| | - Li Xu
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohong Zeng
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Wu
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Transplant Engineering and Immunology NHC, West China Hospital, Sichuan University, Chengdu, China
| | - Faqing Liang
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Lv
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Zhenggui Du, ; Qing Lv,
| | - Zhenggui Du
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Zhenggui Du, ; Qing Lv,
| |
Collapse
|
14
|
Maharaj K, Uriepero A, Sahakian E, Pinilla-Ibarz J. Regulatory T cells (Tregs) in lymphoid malignancies and the impact of novel therapies. Front Immunol 2022; 13:943354. [PMID: 35979372 PMCID: PMC9376239 DOI: 10.3389/fimmu.2022.943354] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022] Open
Abstract
Regulatory T cells (Tregs) are responsible for maintaining immune homeostasis by controlling immune responses. They can be characterized by concomitant expression of FoxP3, CD25 and inhibitory receptors such as PD-1 and CTLA-4. Tregs are key players in preventing autoimmunity and are dysregulated in cancer, where they facilitate tumor immune escape. B-cell lymphoid malignancies are a group of diseases with heterogenous molecular characteristics and clinical course. Treg levels are increased in patients with B-cell lymphoid malignancies and correlate with clinical outcomes. In this review, we discuss studies investigating Treg immunobiology in B-cell lymphoid malignancies, focusing on clinical correlations, mechanisms of accumulation, phenotype, and function. Overarching trends suggest that Tregs can be induced directly by tumor cells and recruited to the tumor microenvironment where they suppress antitumor immunity to facilitate disease progression. Further, we highlight studies showing that Tregs can be modulated by novel therapeutic agents such as immune checkpoint blockade and targeted therapies. Treg disruption by novel therapeutics may beneficially restore immune competence but has been associated with occurrence of adverse events. Strategies to achieve balance between these two outcomes will be paramount in the future to improve therapeutic efficacy and safety.
Collapse
Affiliation(s)
- Kamira Maharaj
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Angimar Uriepero
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Eva Sahakian
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Javier Pinilla-Ibarz
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- *Correspondence: Javier Pinilla-Ibarz,
| |
Collapse
|
15
|
Zhou X, Ren T, Zan H, Hua C, Guo X. Novel Immune Checkpoints in Esophageal Cancer: From Biomarkers to Therapeutic Targets. Front Immunol 2022; 13:864202. [PMID: 35669786 PMCID: PMC9163322 DOI: 10.3389/fimmu.2022.864202] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/25/2022] [Indexed: 12/24/2022] Open
Abstract
Esophageal cancer ranks as the sixth most common cause of cancer death worldwide. Due to the limited efficacy of conventional therapeutic strategies, including surgery, chemotherapy, and radiotherapy, treatments are still far from satisfactory in terms of survival, prompting the search for novel treatment methods. Immune checkpoints play crucial roles in immune evasion mediated by tumor cells, and successful clinical outcomes have been achieved via blocking these pathways. However, only a small fraction of patients can benefit from current immune checkpoint inhibitors targeting programmed cell death ligand-1 (PD-L1) and cytotoxic T-lymphocyte-associated protein-4. Unfortunately, some patients show primary and/or acquired resistance to immune checkpoint inhibitors. Until now, novel immune checkpoint pathways have rarely been studied in esophageal cancer, and there is a great need for biomarkers to predict who will benefit from existing strategies. Herein, we primarily discuss the roles of new immune checkpoints as predictive biomarkers and therapeutic targets for esophageal cancer. In addition, we summarize the ongoing clinical trials and provide future research directions targeting these pathways.
Collapse
Affiliation(s)
- Xueyin Zhou
- School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ting Ren
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hongyuan Zan
- School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Chunyan Hua, ; Xufeng Guo,
| | - Xufeng Guo
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Chunyan Hua, ; Xufeng Guo,
| |
Collapse
|
16
|
Tumor Immune Microenvironment in Lymphoma: Focus on Epigenetics. Cancers (Basel) 2022; 14:cancers14061469. [PMID: 35326620 PMCID: PMC8946119 DOI: 10.3390/cancers14061469] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/23/2022] [Accepted: 03/11/2022] [Indexed: 02/06/2023] Open
Abstract
Lymphoma is a neoplasm arising from B or T lymphocytes or natural killer cells characterized by clonal lymphoproliferation. This tumor comprises a diverse and heterogeneous group of malignancies with distinct clinical, histopathological, and molecular characteristics. Despite advances in lymphoma treatment, clinical outcomes of patients with relapsed or refractory disease remain poor. Thus, a deeper understanding of molecular pathogenesis and tumor progression of lymphoma is required. Epigenetic alterations contribute to cancer initiation, progression, and drug resistance. In fact, over the past decade, dysregulation of epigenetic mechanisms has been identified in lymphomas, and the knowledge of the epigenetic aberrations has led to the emergence of the promising epigenetic therapy field in lymphoma tumors. However, epigenetic aberrations in lymphoma not only have been found in tumor cells, but also in cells from the tumor microenvironment, such as immune cells. Whereas the epigenetic dysregulation in lymphoma cells is being intensively investigated, there are limited studies regarding the epigenetic mechanisms that affect the functions of immune cells from the tumor microenvironment in lymphoma. Therefore, this review tries to provide a general overview of epigenetic alterations that affect both lymphoma cells and infiltrating immune cells within the tumor, as well as the epigenetic cross-talk between them.
Collapse
|
17
|
Xu Y, Zhou X, Zhang S, Nanding A, Xuan Q. Expression and Prognostic Value of Glucose Transporter 3 in Diffuse Large B Cell Lymphoma. Onco Targets Ther 2022; 15:181-191. [PMID: 35250277 PMCID: PMC8888198 DOI: 10.2147/ott.s338826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 02/09/2022] [Indexed: 12/17/2022] Open
Abstract
Background Several reports have suggested that glucose transporter 3 (GLUT-3) promotes tumor metastasis. The aim of this study was to examine the relationship between the expression level of GLUT-3 and the prognosis of patients with diffuse large B cell lymphoma (DLBCL). Methods The GLUT-3 expression levels in 91 DLBCL patients were evaluated by immunohistochemistry. The relationships between GLUT-3 expression level and clinicopathological characteristics and progression-free survival (PFS) of DLBCL patients were analyzed. The use of validation cohorts confirmed the predictive value of GLUT-3 expression. The correlation between GLUT-3 and immune cell infiltration was investigated using the Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts system and the analysis of the infiltrating score was obtained by single sample Gene Set Enrichment Analysis. Results Expression of GLUT-3, which is highly expressed in DLBCL patients, was significantly associated with elevated serum LDH level, recurrence and Ki-67 status. Kaplan–Meier analysis showed that high GLUT-3 expression levels in DLBCL were related to poor PFS. Univariate and multivariate analyses results showed that low GLUT-3 expression level was significantly but independently associated with favorable PFS in DLBCL patients. GLUT-3 expression was also correlated with immune cell infiltration and the analysis of the infiltrating score. Conclusion Our results indicate that GLUT-3 may act as a potential independent prognostic factor in DLBCL patients. The difference of the immune microenvironment in DLBCL patients may be predicted by the expression level of GLUT-3.
Collapse
Affiliation(s)
- Yongpeng Xu
- Department of Urology Surgery, The Fourth Affiliated Hospital Zhejiang University School of Medicine, Yiwu, Zhejiang Province, 322000, People’s Republic of China
| | - Xinglu Zhou
- Department of PET/CT Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150081, People’s Republic of China
| | - Shuai Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150081, People’s Republic of China
| | - Abiyasi Nanding
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150081, People’s Republic of China
| | - Qijia Xuan
- Department of Medical Oncology, The Fourth Affiliated Hospital Zhejiang University School of Medicine, Yiwu, Zhejiang Province, 322000, People’s Republic of China
- Correspondence: Qijia Xuan, Department of Medical Oncology, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, 322000, People’s Republic of China, Tel +86-579-582303, Email
| |
Collapse
|
18
|
Tian T, Li Z. Targeting Tim-3 in Cancer With Resistance to PD-1/PD-L1 Blockade. Front Oncol 2021; 11:731175. [PMID: 34631560 PMCID: PMC8492972 DOI: 10.3389/fonc.2021.731175] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022] Open
Abstract
Programmed death receptor 1 (PD-1) or programmed death ligand 1 (PD-L1) blocking therapy has completely changed the treatment pattern of malignant tumors. It has been tested in a wide range of malignant tumors and achieved clinical success. It might be a promising cancer treatment strategy. However, one of the important disadvantages of PD-1/PD-L1 blocking therapy is that only a few patients have a positive response to it. In addition, primary or acquired drug resistance can also lead to cancer recurrence in patients with clinical response. Therefore, it is very important to overcome the resistance of PD-1/PD-L1 blocking therapy and improve the overall response rate of patients to the immunotherapy. T cell immunoglobulin and mucin domain molecule 3 (Tim-3) belongs to the co-inhibitory receptor family involved in immune checkpoint function. Due to adaptive resistance, the expression of Tim-3 is up-regulated in PD-1/PD-L1 blocking therapy resistant tumors. Therefore, blocking the immune checkpoint Tim-3 might antagonize the resistance of PD-1/PD-L1 blocking therapy. This review systematically introduces the preclinical and clinical data of combined blockade of Tim-3 and PD-1/PD-L1 in cancer immunotherapy, and discusses the prospect of overcoming the drug resistance of PD-1/PD-L1 blockade therapy through blockade of Tim-3.
Collapse
Affiliation(s)
- Tian Tian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Zheng X, Li S, Yang H. Roles of Toll-Like Receptor 3 in Human Tumors. Front Immunol 2021; 12:667454. [PMID: 33986756 PMCID: PMC8111175 DOI: 10.3389/fimmu.2021.667454] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 04/06/2021] [Indexed: 12/29/2022] Open
Abstract
Toll-like receptor 3 (TLR3) is an important member of the TLR family, which is an important group of pathogen-associated molecular patterns. TLR3 can recognize double-stranded RNA and induce activation of NF-κB and the production of type I interferons. In addition to its immune-associated role, TLR3 has also been detected in some tumors. However TLR3 can play protumor or antitumor roles in different tumors or cell lines. Here, we review the basic signaling associated with TLR3 and the pro- or antitumor roles of TLR3 in different types of tumors and discuss the possible reasons for the opposing roles of TLR3 in tumors.
Collapse
Affiliation(s)
- Xin Zheng
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Song Li
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hui Yang
- Department of Neurosurgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|