1
|
Gao D. The role of non-malignant B cells in malignant hematologic diseases. Hematology 2025; 30:2466261. [PMID: 39964954 DOI: 10.1080/16078454.2025.2466261] [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: 06/02/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025] Open
Abstract
The tumor microenvironment (TME) represents a heterogeneous, complicated ecosystem characterized by intricate interactions between tumor cells and immune cells. During the past decade, immune cells especially T cells were found to play an important role in the progression of tumor and many related immune checkpoints drugs were created. In recent years, more and more scientists revealed the critical role of B-cells within the TME, particularly various populations of non-malignant B cells. Some studies indicated that non-malignant B cells may exert a 'double-edged sword' role in solid tumors. However, there has been comparatively less focus on the role of non-malignant B cells in hematologic malignancies. In this review, we characterized the development of B cells and summarized its functions of antitumor immunity within TME, with an emphasis on elucidating the roles and potential mechanisms of non-malignant B cells in the progression of hematologic diseases including classical Hodgkin's lymphoma, non-Hodgkin's B-cell lymphoma, non-Hodgkin's T-cell lymphoma, leukemia and multiple myeloma.
Collapse
Affiliation(s)
- Daquan Gao
- Department of Hematology, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, People's Republic of China
| |
Collapse
|
2
|
Attygalle AD, Chak PK, Madej E, Tzioni MM, Chen Z, Du MQ. Multi-neoplastic potentials including metachronous clonally unrelated nodal T-follicular helper cell lymphomas in clonal haematopoiesis. Histopathology 2025; 86:652-656. [PMID: 39526912 DOI: 10.1111/his.15367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Affiliation(s)
- Ayoma D Attygalle
- Department of Histopathology, The Royal Marsden Hospital, London, UK
| | - Pui Kwan Chak
- Department of Histopathology, The Royal Marsden Hospital, London, UK
| | - Ewelina Madej
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Maria-Myrsini Tzioni
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Zi Chen
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ming-Qing Du
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
3
|
Zhu M, Li N, Fan L, Wu R, Cao L, Ren Y, Lu C, Zhang L, Cai Y, Shi Y, Lin Z, Lu X, Leng J, Zhong S, Hu X, Huang B, Huang R, Zhou W, Yao D, Wu L, Wu W, Liu Q, Xia P, Chen R, Shi W, Zhang R, Lv S, Wang C, Yu L, Li J, Wang Q, Li K, Jin H. Single-cell transcriptomic and spatial analysis reveal the immunosuppressive microenvironment in relapsed/refractory angioimmunoblastic T-cell lymphoma. Blood Cancer J 2024; 14:218. [PMID: 39695118 DOI: 10.1038/s41408-024-01199-0] [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: 10/05/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is a kind of aggressive T-cell lymphoma with significant enrichment of non-malignant tumor microenvironment (TME) cells. However, the complexity of TME in AITL progression is poorly understood. We performed single-cell RNA-Seq (scRNA-seq) and imaging mass cytometry (IMC) analysis to compare the cellular composition and spatial architecture between relapsed/refractory AITL (RR-AITL) and newly diagnosed AITL (ND-AITL). Our results showed that the malignant T follicular helper (Tfh) cells showed significantly increased proliferation driven by transcriptional activation of YY1 in RR-AITL, which is markedly associated with the poor prognosis of AITL patients. The CD8+ T cell proportion and cytotoxicity decreased in RR-AITL TME, resulting from elevated expression of the inhibitory checkpoints such as PD-1, TIGIT, and CTLA4. Notably, the transcriptional pattern of B cells in RR-AITL showed an intermediate state of malignant transformation to B-cell-lymphoma, and contributed to immune evasion by highly expressing CD47 and PD-L1. Besides, compared to ND-AITL samples, myeloid-cells-centered spatial communities were more prevalent but showed reduced phagocytic activity and impaired antigen processing and presentation in RR-AITL TME. Furthermore, specific inhibitory ligand-receptor interactions, such as CLEC2D-KLRB1, CTLA4-CD86, and MIF-CD74, were exclusively identified in the RR-AITL TME. Our study provides a high-resolution characterization of the immunosuppression ecosystem and reveals the potential therapeutic targets for RR-AITL patients.
Collapse
Affiliation(s)
- Mengyan Zhu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ning Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu, China
| | - Lei Fan
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Rongrong Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Cao
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Yimin Ren
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Chuanyang Lu
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu, China
| | - Lishen Zhang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Cai
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuzhu Shi
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zihan Lin
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xueying Lu
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Jiayan Leng
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Shiyang Zhong
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xingfei Hu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bin Huang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Runheng Huang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wanting Zhou
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Diru Yao
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lingxiang Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Quanzhong Liu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peng Xia
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ruize Chen
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China
| | - Wenyu Shi
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Ruohan Zhang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sali Lv
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunling Wang
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu, China
| | - Liang Yu
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu, China
| | - Jianyong Li
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China.
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China.
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China.
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China.
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
- Biomedical Big Data Center, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Kening Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China.
- Department of Hematology of the Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huai'an, Jiangsu, China.
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China.
- Biomedical Big Data Center, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Hui Jin
- Lymphoma Center, Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Collaborative Innovation Center for Personalized Cancer Medicine, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Nanjing Medical University, Nanjing, Jiangsu, China.
- Key Laboratory of Hematology of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
Gu X, Li D, Wu P, Zhang C, Cui X, Shang D, Ma R, Liu J, Sun N, He J. Revisiting the CXCL13/CXCR5 axis in the tumor microenvironment in the era of single-cell omics: Implications for immunotherapy. Cancer Lett 2024; 605:217278. [PMID: 39332588 DOI: 10.1016/j.canlet.2024.217278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/22/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
As one of the important members of the family of chemokines and their receptors, the CXCL13/CXCR5 axis is involved in follicle formation in normal lymphoid tissues and the establishment of somatic cavity immunity under physiological conditions, as well as being associated with a wide range of infectious, autoimmune, and tumoral diseases. Here in this review, we focus on its role in tumors. Traditional studies have found the axis to be both pro- and anti-tumorigenic, involving a variety of immune cells, including the tumor cells themselves and those in the tumor microenvironment (TME), and the prognostic significance of this axis is clinical context-dependent. With the development of techniques at the single-cell level, we were able to explain in detail the status of the CXCL13/CXCR5 axis in the TME based on real clinical samples and found that it involves a range of crucial intrinsic anti-tumor immune processes in the TME and is therefore important in tumor immunotherapy. We summarize the cellular subsets, physiological functions, and prognostic significance associated with this axis in the most promising immune checkpoint inhibitor (ICI) therapies of the day and summarize possible therapeutic ideas based on this axis. As with any TME study, the most important takeaway is that the complexity of the CXCL13/CXCR5 axis in TME suggests the importance of personalized therapy in tumor therapy.
Collapse
Affiliation(s)
- Xuanyu Gu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dongyu Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Wu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chaoqi Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinyu Cui
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dexin Shang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China; 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ruijie Ma
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jingjing Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
5
|
Sainz TP, Sahu V, Gomez JA, Dcunha NJ, Basi AV, Kettlun C, Sarami I, Burks JK, Sampath D, Vega F. Role of the Crosstalk B:Neoplastic T Follicular Helper Cells in the Pathobiology of Nodal T Follicular Helper Cell Lymphomas. J Transl Med 2024; 104:102147. [PMID: 39389311 DOI: 10.1016/j.labinv.2024.102147] [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: 06/20/2024] [Revised: 09/06/2024] [Accepted: 09/29/2024] [Indexed: 10/12/2024] Open
Abstract
Angioimmunoblastic T-cell lymphoma (AITL), the most common form of peripheral T-cell lymphoma, originates from follicular helper T (Tfh) cells and is notably resistant to current treatments. The disease progression and maintenance, at least in early stages, are driven by a complex interplay between neoplastic Tfh and clusters of B-cells within the tumor microenvironment, mirroring the functional crosstalk observed inside germinal centers. This interaction is further complicated by recurrent mutations, such as TET2 and DNMT3A, which are present in both Tfh cells and B-cells. These findings suggest that the symbiotic relationship between these 2 cell types could represent a therapeutic vulnerability. This review examines the key components and signaling mechanisms involved in the synapses between B-cells and Tfh cells, emphasizing their significant role in the pathobiology of AITL and potential as therapeutic targets.
Collapse
Affiliation(s)
- Tania P Sainz
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Vishal Sahu
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Javier A Gomez
- Department of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Nicholas J Dcunha
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - Akshay V Basi
- Department of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Claudia Kettlun
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Iman Sarami
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Jared K Burks
- Department of Leukemia, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Deepa Sampath
- Hematopoietic Biology and Malignancy, MD Anderson Cancer Center, The University of Texas, Houston, Texas
| | - Francisco Vega
- Department of Hematopathology, MD Anderson Cancer Center, The University of Texas, Houston, Texas.
| |
Collapse
|
6
|
Lu C, Li M, Fu J, Fan X, Zhong L, Li Y, Xi Q. cyTRBC1 evaluation rapidly identifies sCD3-negative peripheral T-cell lymphomas and reveals a novel type of sCD3-negative T-cell clone with uncertain significance. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2024; 106:465-475. [PMID: 38818861 DOI: 10.1002/cyto.b.22182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/19/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024]
Abstract
The flow cytometry-based evaluation of TRBC1 expression has been demonstrated as a rapid and specific method for detecting T-cell clones in sCD3-positive TCRαβ+ mature T-cell lymphoma. The aim of the study was to validate the utility of surface (s) TRBC1 and cytoplastic (cy) TRBC1 assessment in detecting clonality of sCD3-negative peripheral T-cell lymphomas (PTCLs), as well as exploring the existence and characteristics of sCD3-negative clonal T-cell populations with uncertain significance (T-CUS). Evaluation of sTRBC1 and cyTRBC1 were assessed on 61 samples from 37 patients with sCD3-negative PTCLs, including 26 angioimmunoblastic T-cell lymphoma (AITL) patients and 11 non-AITL patients. The sCD3-negative T-CUS were screened from 1602 patients without T-cell malignancy and 100 healthy individuals. Additionally, the clonality of cells was further detected through T-cell gene rearrangement analysis. We demonstrated the monotypic expression patterns of cyTRBC1 in all sCD3-negative PTCLs. Utilizing the cyTRBC1 evaluation assay, we identified a novel and rare subtype of sCD3-negative T-CUS for the first time among 13 out of 1602 (0.8%) patients without T-cell malignancy. The clonality of these cells was further confirmed through T-cell gene rearrangement analysis. This subset exhibited characteristics such as sCD3-cyCD3 + CD4 + CD45RO+, closely resembling AITL rather than non-AITL. Further analysis revealed that sCD3-negative T-CUS exhibited a smaller clone size in the lymph node and mass specimens compared to AITL patients. However, the clone size of sCD3-negative T-CUS was significantly lower than that of non-AITL patients in both specimen groups. In conclusion, we validated the diagnostic utility of cyTRBC1 in detecting sCD3-negative T-cell clonality, provided a comprehensive analysis of sCD3-negative T-CUS, and established a framework and provided valuable insights for distinguishing sCD3-negative T-CUS from sCD3-negative PTCLs based on their phenotypic properties and clone size.
Collapse
Affiliation(s)
- Cong Lu
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingyong Li
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Fu
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoming Fan
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ling Zhong
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yanxin Li
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qian Xi
- Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
7
|
Moskowitz AJ, Stuver RN, Horwitz SM. Current and upcoming treatment approaches to common subtypes of PTCL (PTCL, NOS; ALCL; and TFHs). Blood 2024; 144:1887-1897. [PMID: 38306597 PMCID: PMC11830973 DOI: 10.1182/blood.2023021789] [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: 12/05/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/04/2024] Open
Abstract
ABSTRACT The treatment of common nodal peripheral T-cell lymphomas (PTCLs), including PTCL, not otherwise specified (PTCL, NOS), anaplastic large-cell lymphomas, and T-follicular helper lymphomas, is evolving. These entities are currently treated similarly with cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) or cyclophosphamide, doxorubicin, vincristine, etoposide, and prednisone (CHOEP) for CD30-negative diseases, or brentuximab vedotin plus cyclophosphamide, doxorubicin, and prednisone (CHP) for CD30-positive diseases, followed by consolidation with autologous stem cell transplantation in the first remission. Ongoing improvements in PTCL classification, identification of predictive biomarkers, and development of new targeted agents will lead to more specific therapies that address the unique biologic and clinical properties of each entity. For example, widespread efforts focused on molecular profiling of PTCL, NOS is likely to identify distinct subtypes that warrant different treatment approaches. New agents, such as EZH1/2 and JAK/STAT pathway inhibitors, have broadened treatment options for relapsed or refractory diseases. Furthermore, promising strategies for optimizing immune therapy for PTCL are currently under investigation and have the potential to significantly alter the therapeutic landscape. Ongoing frontline study designs incorporate an understanding of disease biology and drug sensitivities and are poised to evaluate whether newer-targeted agents should be incorporated into frontline settings for various disease entities. Although current treatment strategies lump most disease entities together, future treatments will include distinct strategies for each disease subtype that optimize therapy for individuals. This movement toward individualized therapy will ultimately lead to dramatic improvements in the prognosis of patients with PTCL.
Collapse
Affiliation(s)
- Alison J. Moskowitz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert N. Stuver
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Steven M. Horwitz
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
8
|
Vogelsberg A, Harland L, Borgmann V, Otto F, Weller JF, Nann D, Quintanilla-Martinez L, Fend F. Clonal haematopoiesis: A common progenitor for cytotoxic peripheral T-cell lymphoma and angioimmunoblastic T-cell lymphoma. Br J Haematol 2024; 204:2071-2076. [PMID: 38323682 DOI: 10.1111/bjh.19335] [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: 11/26/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/08/2024]
Abstract
Recent studies have shown that follicular helper T-cell lymphoma of angioimmunoblastic type (AITL), the most common nodal peripheral T-cell lymphoma (PTCL), frequently arises in a background of clonal haematopoiesis (CH), a preneoplastic condition affecting up to 40% of elderly individuals. Data on a potential CH association are limited for other PTCL. We report a unique patient who sequentially developed both cytotoxic PTCL, not otherwise specified and AITL with distinct T-cell receptor rearrangements but shared somatic mutations originating from the same CH clone, thus providing convincing evidence that CH can give rise to T-cell neoplasms of different lineage.
Collapse
Affiliation(s)
- Antonio Vogelsberg
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tuebingen, Tuebingen, Germany
| | - Lennart Harland
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tuebingen, Tuebingen, Germany
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Vanessa Borgmann
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tuebingen, Tuebingen, Germany
| | - Franziska Otto
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tuebingen, Tuebingen, Germany
| | - Jan F Weller
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Dominik Nann
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tuebingen, Tuebingen, Germany
| | - Leticia Quintanilla-Martinez
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tuebingen, Tuebingen, Germany
| | - Falko Fend
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tuebingen, Tuebingen, Germany
| |
Collapse
|
9
|
Suma S, Suehara Y, Fujisawa M, Abe Y, Hattori K, Makishima K, Sakamoto T, Sawa A, Bando H, Kaji D, Sugio T, Kato K, Akashi K, Matsue K, Carreras J, Nakamura N, Suzuki A, Suzuki Y, Ito K, Shiiba H, Chiba S, Sakata-Yanagimoto M. Tumor heterogeneity and immune-evasive T follicular cell lymphoma phenotypes at single-cell resolution. Leukemia 2024; 38:340-350. [PMID: 38012392 PMCID: PMC10844096 DOI: 10.1038/s41375-023-02093-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023]
Abstract
T follicular helper (TFH) cell lymphomas (TFHLs) are characterized by TFH-like properties and accompanied by substantial immune-cell infiltration into tumor tissues. Nevertheless, the comprehensive understanding of tumor-cell heterogeneity and immune profiles of TFHL remains elusive. To address this, we conducted single-cell transcriptomic analysis on 9 lymph node (LN) and 16 peripheral blood (PB) samples from TFHL patients. Tumor cells were divided into 5 distinct subclusters, with significant heterogeneity observed in the expression levels of TFH markers. Copy number variation (CNV) and trajectory analyses indicated that the accumulation of CNVs, together with gene mutations, may drive the clonal evolution of tumor cells towards TFH-like and cell proliferation phenotypes. Additionally, we identified a novel tumor-cell-specific marker, PLS3. Notably, we found a significant increase in exhausted CD8+ T cells with oligoclonal expansion in TFHL LNs and PB, along with distinctive immune evasion characteristics exhibited by infiltrating regulatory T, myeloid, B, and natural killer cells. Finally, in-silico and spatial cell-cell interaction analyses revealed complex networking between tumor and immune cells, driving the formation of an immunosuppressive microenvironment. These findings highlight the remarkable tumor-cell heterogeneity and immunoevasion in TFHL beyond previous expectations, suggesting potential roles in treatment resistance.
Collapse
Affiliation(s)
- Sakurako Suma
- Department of Hematology, University of Tsukuba Hospital, Tsukuba, Japan
| | - Yasuhito Suehara
- Department of Hematology, University of Tsukuba Hospital, Tsukuba, Japan
| | - Manabu Fujisawa
- Department of Hematology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Centre for Lymphoid Cancer, BC Cancer, Vancouver, BC, Canada
| | - Yoshiaki Abe
- Department of Hematology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Keiichiro Hattori
- Department of Hematology, University of Tsukuba Hospital, Tsukuba, Japan
- Department of Hematology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kenichi Makishima
- Department of Hematology, University of Tsukuba Hospital, Tsukuba, Japan
| | - Tatsuhiro Sakamoto
- Department of Hematology, University of Tsukuba Hospital, Tsukuba, Japan
- Department of Hematology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Aya Sawa
- Department of Breast-Thyroid-Endocrine Surgery, University of Tsukuba Hospital, Tsukuba, Japan
| | - Hiroko Bando
- Department of Breast-Thyroid-Endocrine Surgery, University of Tsukuba Hospital, Tsukuba, Japan
| | - Daisuke Kaji
- Department of Hematology, Toranomon Hospital, Tokyo, Japan
| | - Takeshi Sugio
- Department of Medicine, Division of Oncology, Stanford University, Stanford, CA, USA
| | - Koji Kato
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Kosei Matsue
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Japan
| | - Joaquim Carreras
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan
| | - Naoya Nakamura
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan
| | - Ayako Suzuki
- Department of Computational Biology and Medical Sciences, the University of Tokyo, Kashiwa, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, the University of Tokyo, Kashiwa, Japan
| | - Ken Ito
- Oncology Business Unit, Eisai Co., Ltd., Tsukuba, Japan
| | - Hiroyuki Shiiba
- Oncology Department, Medical Head Quarters, Eisai Co., Ltd., Tokyo, Japan
| | - Shigeru Chiba
- Department of Hematology, University of Tsukuba Hospital, Tsukuba, Japan
- Department of Hematology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Mamiko Sakata-Yanagimoto
- Department of Hematology, University of Tsukuba Hospital, Tsukuba, Japan.
- Department of Hematology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan.
- Division of Advanced Hemato-Oncology, Transborder Medical Research Center, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
10
|
Zhang Q, Yin L, Lai Q, Zhao Y, Peng H. Advances in the pathogenesis and therapeutic strategies of angioimmunoblastic T-cell lymphoma. Clin Exp Med 2023; 23:4219-4235. [PMID: 37759042 DOI: 10.1007/s10238-023-01197-9] [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: 07/08/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023]
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is an aggressive subtype of peripheral T-cell lymphomas with its cell origin determined to be follicular helper T-cells. AITL is characterized by a prominent tumor microenvironment involving dysregulation of immune cells, signaling pathways, and extracellular matrix. Significant progress has been made in the molecular pathophysiology of AITL, including genetic mutations, immune metabolism, hematopoietic-derived microenvironment, and non-hematopoietic microenvironment cells. Early diagnosis, detection of severe complications, and timely effective treatment are crucial for managing AITL. Treatment typically involves various combination chemotherapies, but the prognosis is often poor, and relapsed and refractory AITL remains challenging, necessitating improved treatment strategies. Therefore, this article provides an overview of the pathogenesis and latest advances in the treatment of AITL, with a focus on potential therapeutic targets, novel treatment strategies, and emerging immunotherapeutic approaches.
Collapse
Affiliation(s)
- Qingyang Zhang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Le Yin
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qinqiao Lai
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yan Zhao
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, 410011, Hunan, China.
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, 410011, Hunan, China.
| |
Collapse
|
11
|
Zhu Q, Yang Y, Deng X, Chao N, Chen Z, Ye Y, Zhang W, Liu W, Zhao S. High CD8 +tumor-infiltrating lymphocytes indicate severe exhaustion and poor prognosis in angioimmunoblastic T-cell lymphoma. Front Immunol 2023; 14:1228004. [PMID: 37781365 PMCID: PMC10540231 DOI: 10.3389/fimmu.2023.1228004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/23/2023] [Indexed: 10/03/2023] Open
Abstract
Background Exhaustion of CD8+ tumor-infiltrating lymphocytes (TILs), characterized by the overexpression of immune checkpoints (IC), is a major impediment to anti-tumor immunity. However, the exhaustion status of CD8+TILs in angioimmunoblastic T cell lymphoma (AITL) remains unclear. Therefore, we aimed to elucidate the exhaustion status of CD8+TILs in AITL and its influence on prognosis. Methods The correlation between CD8+TILs and IC expression in AITL was analyzed using single-cell RNA sequencing (n = 2), flow cytometry (n = 20), and RNA sequencing (n = 20). Biological changes related to CD8+TILs exhaustion at different cytotoxic T lymphocyte (CTL) levels (mean expression levels of CD8A, CD8B, GZMA, GZMB, and PRF1) in AITL were evaluated using RNA sequencing (n = 20) and further validated using the GEO dataset (n = 51). The impact of CD8 protein expression and CTL levels on patient prognosis was analyzed using flow cytometry and RNA sequencing, respectively. Results Our findings demonstrated that the higher the infiltration of CD8+TILs, the higher was the proportion of exhausted CD8+TILs characterized by the overexpression of multiple IC. This was accompanied by extensive exhaustion-related biological changes, which suggested severe exhaustion in CD8+TILs and may be one of the main reasons for the poor prognosis of patients with high CD8+TILs and CTL. Conclusion Our study comprehensively reveals the exhaustion status of CD8+TILs and their potential negative impact on AITL prognosis, which facilitates further mechanistic studies and is valuable for guiding immunotherapy strategies.
Collapse
Affiliation(s)
- Qiqi Zhu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yiming Yang
- Department of Pathology, North Sichuan Medical College, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Xueqin Deng
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Ningning Chao
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Zihang Chen
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Yunxia Ye
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Wenyan Zhang
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Weiping Liu
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| | - Sha Zhao
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Yadav M, Uikey BN, Rathore SS, Gupta P, Kashyap D, Kumar C, Shukla D, Vijayamahantesh, Chandel AS, Ahirwar B, Singh AK, Suman SS, Priyadarshi A, Amit A. Role of cytokine in malignant T-cell metabolism and subsequent alternation in T-cell tumor microenvironment. Front Oncol 2023; 13:1235711. [PMID: 37746258 PMCID: PMC10513393 DOI: 10.3389/fonc.2023.1235711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
T cells are an important component of adaptive immunity and T-cell-derived lymphomas are very complex due to many functional sub-types and functional elasticity of T-cells. As with other tumors, tissues specific factors are crucial in the development of T-cell lymphomas. In addition to neoplastic cells, T- cell lymphomas consist of a tumor micro-environment composed of normal cells and stroma. Numerous studies established the qualitative and quantitative differences between the tumor microenvironment and normal cell surroundings. Interaction between the various component of the tumor microenvironment is crucial since tumor cells can change the microenvironment and vice versa. In normal T-cell development, T-cells must respond to various stimulants deferentially and during these courses of adaptation. T-cells undergo various metabolic alterations. From the stage of quiescence to attention of fully active form T-cells undergoes various stage in terms of metabolic activity. Predominantly quiescent T-cells have ATP-generating metabolism while during the proliferative stage, their metabolism tilted towards the growth-promoting pathways. In addition to this, a functionally different subset of T-cells requires to activate the different metabolic pathways, and consequently, this regulation of the metabolic pathway control activation and function of T-cells. So, it is obvious that dynamic, and well-regulated metabolic pathways are important for the normal functioning of T-cells and their interaction with the microenvironment. There are various cell signaling mechanisms of metabolism are involved in this regulation and more and more studies have suggested the involvement of additional signaling in the development of the overall metabolic phenotype of T cells. These important signaling mediators include cytokines and hormones. The impact and role of these mediators especially the cytokines on the interplay between T-cell metabolism and the interaction of T-cells with their micro-environments in the context of T-cells lymphomas are discussed in this review article.
Collapse
Affiliation(s)
- Megha Yadav
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Blessi N. Uikey
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Priyanka Gupta
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Diksha Kashyap
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Chanchal Kumar
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Dhananjay Shukla
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Vijayamahantesh
- Department of Immunology and Microbiology, University of Missouri, Columbia, SC, United States
| | - Arvind Singh Chandel
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo, Japan
| | - Bharti Ahirwar
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Shashi Shekhar Suman
- Department of Zoology, Udayana Charya (UR) College, Lalit Narayan Mithila University, Darbhanga, India
| | - Amit Priyadarshi
- Department of Zoology, Veer Kunwar Singh University, Arrah, India
| | - Ajay Amit
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| |
Collapse
|
13
|
Huang D, Ma N, Li X, Gou Y, Duan Y, Liu B, Xia J, Zhao X, Wang X, Li Q, Rao J, Zhang X. Advances in single-cell RNA sequencing and its applications in cancer research. J Hematol Oncol 2023; 16:98. [PMID: 37612741 PMCID: PMC10463514 DOI: 10.1186/s13045-023-01494-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/09/2023] [Indexed: 08/25/2023] Open
Abstract
Cancers are a group of heterogeneous diseases characterized by the acquisition of functional capabilities during the transition from a normal to a neoplastic state. Powerful experimental and computational tools can be applied to elucidate the mechanisms of occurrence, progression, metastasis, and drug resistance; however, challenges remain. Bulk RNA sequencing techniques only reflect the average gene expression in a sample, making it difficult to understand tumor heterogeneity and the tumor microenvironment. The emergence and development of single-cell RNA sequencing (scRNA-seq) technologies have provided opportunities to understand subtle changes in tumor biology by identifying distinct cell subpopulations, dissecting the tumor microenvironment, and characterizing cellular genomic mutations. Recently, scRNA-seq technology has been increasingly used in cancer studies to explore tumor heterogeneity and the tumor microenvironment, which has increased the understanding of tumorigenesis and evolution. This review summarizes the basic processes and development of scRNA-seq technologies and their increasing applications in cancer research and clinical practice.
Collapse
Affiliation(s)
- Dezhi Huang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Naya Ma
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Xinlei Li
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Yang Gou
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Yishuo Duan
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Bangdong Liu
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Jing Xia
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Xianlan Zhao
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Xiaoqi Wang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China
- Jinfeng Laboratory, Chongqing, 401329, China
| | - Qiong Li
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
| | - Jun Rao
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
14
|
Zhu S, Zhao Y, Xing C, Guo W, Huang Z, Zhang H, Yin L, Ruan X, Li H, Cheng Z, Wang Z, Peng H. Immune infiltration and drug specificity analysis of different subtypes based on functional status in angioimmunoblastic T-cell lymphoma. Heliyon 2023; 9:e18836. [PMID: 37576233 PMCID: PMC10412840 DOI: 10.1016/j.heliyon.2023.e18836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 07/26/2023] [Accepted: 07/31/2023] [Indexed: 08/15/2023] Open
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is a subtype of peripheral T-cell lymphoma (PTCL) strongly correlated with worse clinical outcomes. However, the role of characteristic pathway-related genes in patients with AITL (e.g., subtype typing and pathogenesis) remains unknown. In this study, we intended to understand the potential role and prognostic value of characteristic pathways in AITL and identified a model for subtype identification based on pathway-related functional status. Transcriptomic (RNA-seq) data were obtained from the Gene Expression Omnibus database for three sets of tumor tissues from AITL patients. AITL was divided into three clusters based on the pathway profile of patients and the best clustering k = 3, and differentially expressed genes (DEGs) in the three clusters were analyzed. The top 45 important variables associated with characteristic pathways, such as Huntington's disease, VEGF signaling pathway, nucleotide excision repair, ubiquitin-mediated proteolysis, purine metabolism, olfactory transduction, etc., were used to construct a subtype identification model. The model was experimentally validated and proved to possess good predictive efficacy. In addition, pathway-related subtype typing was significantly associated with different immune cell infiltration in AITL. Further analysis revealed that the drug IC50 values predicted also differed markedly among the different subtypes, thus further identifying some subtype-specific drugs. Our study indicates a potential role of characteristic pathways in AITL staging for the first time, provides novel insights for future research targeting AITL, and points to potential therapeutic options for patients with different subtypes of AITL.
Collapse
Affiliation(s)
- Shicong Zhu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Yan Zhao
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Cheng Xing
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Wancheng Guo
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Zineng Huang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Huifang Zhang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Le Yin
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Xueqin Ruan
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Heng Li
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Zhao Cheng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Zhihua Wang
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Institute of Molecular Hematology, Central South University, Changsha, Hunan, China
- Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan 410011, China
| |
Collapse
|
15
|
Macapagal SC, Bennani NN. Nodal peripheral T-cell lymphoma: Chemotherapy-free management, are we there yet? Blood Rev 2023; 60:101071. [PMID: 36898933 DOI: 10.1016/j.blre.2023.101071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Peripheral T-cell lymphomas (PTCLs) are a diverse and uncommon type of lymphoid malignancies with a dismal prognosis. Recent advances in genomic studies have shown recurring mutations that are changing our knowledge of the disease's molecular genetics and pathogenesis. As such, new targeted therapies and treatments to improve disease outcomes are currently being explored. In this review, we discussed the current understanding of the nodal PTCL biology with potential therapeutic implications and gave our insights on the promising novel therapies that are currently under study such as immunotherapy, chimeric antigen receptor T-cell therapy, and oncolytic virotherapy.
Collapse
Affiliation(s)
| | - N Nora Bennani
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
16
|
Godfrey J, Chen X, Sunseri N, Cooper A, Yu J, Varlamova A, Zarubin D, Popov Y, Jacobson C, Postovalova E, Xiang Z, Nomie K, Bagaev A, Venkataraman G, Zha Y, Tumuluru S, Smith SM, Kline JP. TIGIT is a key inhibitory checkpoint receptor in lymphoma. J Immunother Cancer 2023; 11:e006582. [PMID: 37364933 PMCID: PMC10410806 DOI: 10.1136/jitc-2022-006582] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND PD-1 checkpoint blockade therapy (CBT) has greatly benefited patients with select solid tumors and lymphomas but has limited efficacy against diffuse large B-cell lymphoma (DLBCL). Because numerous inhibitory checkpoint receptors have been implicated in driving tumor-specific T cell dysfunction, we hypothesized that combinatorial CBT would enhance the activity of anti-PD-1-based therapy in DLBCL. T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) is a coinhibitory receptor expressed on dysfunctional tumor-infiltrating T cells, and TIGIT blockade has demonstrated encouraging activity in combination with PD-1 blockade in murine tumor models and in clinical studies. However, the degree to which TIGIT mediates T cell dysfunction in DLBCL has not been fully explored. RESULTS Here, we demonstrate that TIGIT is broadly expressed on lymphoma-infiltrating T cells (LITs) across a variety of human lymphomas and is frequently coexpressed with PD-1. TIGIT expression is particularly common on LITs in DLBCL, where TIGIT+ LITs often form distinct cellular communities and exhibit significant contact with malignant B cells. TIGIT+/PD-1+ LITs from human DLBCL and murine lymphomas exhibit hypofunctional cytokine production on ex vivo restimulation. In mice with established, syngeneic A20 B-cell lymphomas, TIGIT or PD-1 mono-blockade leads to modest delays in tumor outgrowth, whereas PD-1 and TIGIT co-blockade results in complete rejection of A20 lymphomas in most mice and significantly prolongs survival compared with mice treated with monoblockade therapy. CONCLUSIONS These results provide rationale for clinical investigation of TIGIT and PD-1 blockade in lymphomas, including DLBCL.
Collapse
Affiliation(s)
- James Godfrey
- Hematology, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Xiufen Chen
- Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, USA
| | - Nicole Sunseri
- Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, USA
| | - Alan Cooper
- Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, USA
| | - Jovian Yu
- Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | | | | | | | | | | - Yuanyuan Zha
- Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, USA
| | - Sravya Tumuluru
- Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, USA
| | - Sonali M Smith
- Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, USA
| | - Justin P Kline
- Comprehensive Cancer Center, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
17
|
Zheng J, Wang Z, Pan X, Zhang Z, Li H, Deng X, Liu P, Zhang Q, Na F, Chen C, Niu T, Liu Y. DNMT3A R882H accelerates angioimmunoblastic T-cell lymphoma in mice. Oncogene 2023:10.1038/s41388-023-02699-2. [PMID: 37127775 DOI: 10.1038/s41388-023-02699-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 04/07/2023] [Accepted: 04/18/2023] [Indexed: 05/03/2023]
Abstract
DNA methylation-related genes, including TET2, IDH2, and DNMT3A are highly frequently mutated in angioimmunoblastic T-cell lymphoma (AITL), an aggressive malignancy of T follicular helper (Tfh) cells associated with aberrant immune features. It has been shown that TET2 loss cooperates with RHOAG17V to promote AITL in mice but the functional role of DNMT3A mutations in AITL remains unclear. Here, we report that DNMT3AR882H, the most common mutation of DNMT3A in AITL, accelerates the development of Tet2-/-; RHOAG17V AITL in mice, indicated by the expansion of malignant Tfh cells and aberrant B cells, skin rash, and significantly shortened disease-free survival. To understand the underlying cellular and molecular mechanisms, we performed single-cell transcriptome analyses of lymph nodes of mice transplanted with Tet2-/-, Tet2-/-; RHOAG17V or DNMT3AR882H; Tet2-/-; RHOAG17V hematopoietic stem and progenitor cells. These single-cell landscapes reveal that DNMT3A mutation further activates Tfh cells and leads to rapid and terminal differentiation of B cells, probably through enhancing the interacting PD1/PD-L1, ICOS/ICOSL, CD28/CD86, and ICAM1/ITGAL pairs. Our study establishes the functional roles of DNMT3A mutation in AITL and sheds light on the molecular mechanisms of this disease.
Collapse
Affiliation(s)
- Jianan Zheng
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongwang Wang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangyu Pan
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zhixin Zhang
- Department of Technology, Chengdu ExAb Biotechnology, LTD, Chengdu, China
| | - He Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xintong Deng
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Pengpeng Liu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qi Zhang
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feifei Na
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chong Chen
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China.
| | - Yu Liu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
18
|
Wang S, Sun ST, Zhang XY, Ding HR, Yuan Y, He JJ, Wang MS, Yang B, Li YB. The Evolution of Single-Cell RNA Sequencing Technology and Application: Progress and Perspectives. Int J Mol Sci 2023; 24:ijms24032943. [PMID: 36769267 PMCID: PMC9918030 DOI: 10.3390/ijms24032943] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/01/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
As an emerging sequencing technology, single-cell RNA sequencing (scRNA-Seq) has become a powerful tool for describing cell subpopulation classification and cell heterogeneity by achieving high-throughput and multidimensional analysis of individual cells and circumventing the shortcomings of traditional sequencing for detecting the average transcript level of cell populations. It has been applied to life science and medicine research fields such as tracking dynamic cell differentiation, revealing sensitive effector cells, and key molecular events of diseases. This review focuses on the recent technological innovations in scRNA-Seq, highlighting the latest research results with scRNA-Seq as the core technology in frontier research areas such as embryology, histology, oncology, and immunology. In addition, this review outlines the prospects for its innovative application in traditional Chinese medicine (TCM) research and discusses the key issues currently being addressed by scRNA-Seq and its great potential for exploring disease diagnostic targets and uncovering drug therapeutic targets in combination with multiomics technologies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Bin Yang
- Correspondence: (B.Y.); (Y.-B.L.)
| | - Yu-Bo Li
- Correspondence: (B.Y.); (Y.-B.L.)
| |
Collapse
|
19
|
Cao Q, Liu D, Chen Z, Wang M, Wu M, Zeng G. Upregulated X-C motif chemokine ligand 2 (XCL2) is associated with poor prognosis and increased immune infiltration in clear cell renal cell carcinoma. Cell Signal 2023; 102:110556. [PMID: 36503163 DOI: 10.1016/j.cellsig.2022.110556] [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: 09/26/2022] [Revised: 11/17/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is one of the most popular malignant carcinomas in the genitourinary system. As a novel tumor-related gene, X-C Motif Chemokine Ligand 2 (XCL2) was up-regulated in ccRCC. The current study aims to reveal the functional activity of XCL2 in ccRCC. METHODS The transcriptome profiling, clinical parameters, and simple nucleotide variation profiles of ccRCC samples were obtained from the Cancer Genome Atlas (TCGA) database. The survival analysis, multivariate/univariate Cox analysis, correlation analysis, gene set enrichment analysis (GSEA), and tumor mutation burden (TMB) analysis were performed. Next, immune cell infiltration and immune functions were analyzed. Finally, the functions of XCL2 were investigated in Caki-1 and 786-O cells. RESULTS Upregulated XCL2 was associated with worse overall survival of ccRCC and correlated to age, grade, stage, and T stage. Age, grade, and XCL2 were independent prognostic factors. Significant enrichment in apoptosis, DNA replication, and immune response was demonstrated by GSEA. XCL2 was not only tightly associated with immune cell infiltration, but also significantly linked with several immune functions. Moreover, patients, who had higher XCL2 expression, owned higher levels of TMB. Interestingly, XCL2 was positively correlated with common immune checkpoints. In vitro, XCL2 could inhibit apoptosis, and promote proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of Caki-1 and 786-O cells. CONCLUSIONS In general, the current study suggested that XCL2 may participate in the progression of ccRCC. Importantly, XCL2 may be a potential new target of immunotherapy.
Collapse
Affiliation(s)
- Qingqiong Cao
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Daoquan Liu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhao Chen
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Min Wang
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Meng Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Guang Zeng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
20
|
Terzi di Bergamo L, Guidetti F, Rossi D, Bertoni F, Cascione L. HTGQC and shinyHTGQC: an R package and shinyR application for quality controls of HTG EDGE-seq protocols. GIGABYTE 2022; 2022:gigabyte74. [PMID: 36950141 PMCID: PMC10027062 DOI: 10.46471/gigabyte.74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/01/2022] [Indexed: 12/03/2022] Open
Abstract
Extraction-free HTG EdgeSeq protocols are used to profile sets of genes and measure their expression. Thus, these protocols are frequently used to characterise tumours and their microenvironments. However, although positive and control genes are provided, little indication is given concerning the assessment of the technical success of each sample within the sequencing run. We developed HTGQC, an R package for the quality control of HTG EdgeSeq protocols. Additionally, shinyHTGQC is a shiny application for users without computing knowledge, providing an easy-to-use interface for data quality control and visualisation. Quality checks can be performed on the raw sequencing outputs, and samples are flagged as FAIL or ALERT based on the expression levels of the positive and negative control genes. Availability & Implementation The code is freely available at https://github.com/LodovicoTerzi/HTGQC (R package) and https://lodovico.shinyapps.io/shinyHTGQC/ (shiny application), including test datasets.
Collapse
Affiliation(s)
- Lodovico Terzi di Bergamo
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
- Bioinformatics Core Unit, Swiss Institute of Bioinformatics, Bellinzona, Switzerland
- Department of Health Science and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zurich, Switzerland
| | - Francesca Guidetti
- Lymphoma Genomics unit, Institute of Oncology Research, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Davide Rossi
- Laboratory of Experimental Hematology, Institute of Oncology Research, Bellinzona, Switzerland
| | - Francesco Bertoni
- Lymphoma Genomics unit, Institute of Oncology Research, Bellinzona, Switzerland
| | - Luciano Cascione
- Bioinformatics Core Unit, Swiss Institute of Bioinformatics, Bellinzona, Switzerland
- Lymphoma Genomics unit, Institute of Oncology Research, Bellinzona, Switzerland
| |
Collapse
|
21
|
Fujisawa M, Nguyen TB, Abe Y, Suehara Y, Fukumoto K, Suma S, Makishima K, Kaneko C, Nguyen YT, Usuki K, Narita K, Matsue K, Nakamura N, Ishikawa S, Miura F, Ito T, Suzuki A, Suzuki Y, Mizuno S, Takahashi S, Chiba S, Sakata-Yanagimoto M. Clonal germinal center B cells function as a niche for T-cell lymphoma. Blood 2022; 140:1937-1950. [PMID: 35921527 PMCID: PMC10653021 DOI: 10.1182/blood.2022015451] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 07/13/2022] [Indexed: 11/20/2022] Open
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is proposed to be initiated by age-related clonal hematopoiesis (ACH) with TET2 mutations, whereas the G17V RHOA mutation in immature cells with TET2 mutations promotes the development of T follicular helper (TFH)-like tumor cells. Here, we investigated the mechanism by which TET2-mutant immune cells enable AITL development using mouse models and human samples. Among the 2 mouse models, mice lacking Tet2 in all the blood cells (Mx-Cre × Tet2flox/flox × G17V RHOA transgenic mice) spontaneously developed AITL for approximately up to a year, while mice lacking Tet2 only in the T cells (Cd4-Cre × Tet2flox/flox × G17V RHOA transgenic mice) did not. Therefore, Tet2-deficient immune cells function as a niche for AITL development. Single-cell RNA-sequencing (scRNA-seq) of >50 000 cells from mouse and human AITL samples revealed significant expansion of aberrant B cells, exhibiting properties of activating light zone (LZ)-like and proliferative dark zone (DZ)-like germinal center B (GCB) cells. The GCB cells in AITL clonally evolved with recurrent mutations in genes related to core histones. In silico network analysis using scRNA-seq data identified Cd40-Cd40lg as a possible mediator of GCB and tumor cell cluster interactions. Treatment of AITL model mice with anti-Cd40lg inhibitory antibody prolonged survival. The genes expressed in aberrantly expanded GCB cells in murine tumors were also broadly expressed in the B-lineage cells of TET2-mutant human AITL. Therefore, ACH-derived GCB cells could undergo independent clonal evolution and support the tumorigenesis in AITL via the CD40-CD40LG axis.
Collapse
Affiliation(s)
- Manabu Fujisawa
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Tran B. Nguyen
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshiaki Abe
- Department of Hematology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Yasuhito Suehara
- Department of Hematology, University of Tsukuba Hospital, University of Tsukuba, Tsukuba, Japan
| | - Kota Fukumoto
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Hematology, University of Tsukuba Hospital, University of Tsukuba, Tsukuba, Japan
| | - Sakurako Suma
- Department of Hematology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Kenichi Makishima
- Department of Hematology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Chihiro Kaneko
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yen T.M. Nguyen
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kensuke Usuki
- Department of Hematology, NTT Medical Center Tokyo, Tokyo, Japan
| | - Kentaro Narita
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Japan
| | - Kosei Matsue
- Division of Hematology/Oncology, Department of Internal Medicine, Kameda Medical Center, Kamogawa, Japan
| | - Naoya Nakamura
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Fumihito Miura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Takashi Ito
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Ayako Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba, Japan
| | - Shigeru Chiba
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Hematology, University of Tsukuba Hospital, University of Tsukuba, Tsukuba, Japan
| | - Mamiko Sakata-Yanagimoto
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Department of Hematology, University of Tsukuba Hospital, University of Tsukuba, Tsukuba, Japan
- Division of Advanced Hemato-Oncology, Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
22
|
Chen Z, Zhu Q, Deng X, Yao W, Zhang W, Liu W, Tang Y, Zhao S. Angioimmunoblastic T-cell lymphoma with predominant CD8+ tumor-infiltrating T-cells is a distinct immune pattern with an immunosuppressive microenvironment. Front Immunol 2022; 13:987227. [PMID: 36325319 PMCID: PMC9618886 DOI: 10.3389/fimmu.2022.987227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/27/2022] [Indexed: 11/19/2022] Open
Abstract
Background Angioimmunoblastic T-cell lymphoma (AITL) has a rich tumor microenvironment (TME) that typically harbors plenty of CD4+tumor infiltrating lymphocytes, (TIL)-T-cells (so called common AITL). Nonetheless, AITL with large numbers of CD8+TIL-Ts that outnumber CD4+cells have been observed (CD8-predominant AITL). However, detailed comparison of CD8-predominant AITL and common AITL are still lacking. Methods We compared clinicopathological features, TIL subsets, TME T cell receptor-β (TRB), and immunoglobulin heavy chain (IGH) repertoires, and gene expression profiles in six CD8-predominant and 12 common AITLs using case-control matching (2014 to 2019). Results Comparing with common AITLs, CD8-predominant AITLs showed more frequent edema (P = 0.011), effusion (P = 0.026), high elevated plasma EBV-DNA (P = 0.008), and shorter survival (P = 0.034). Moreover, they had more pronounced eosinophil increase (P = 0.004) and a higher Ki67 index (P = 0.041). Flow cytometry revealed an inverted CD4/CD8 ratio in TIL-Ts and lower TIL-B proportions (P = 0.041). TRB repertoire metrics deteriorated, including lower productive clones (P = 0.014) and higher clonality score (P = 0.019). The IGH repertoire was also narrowed, showing a higher proportion of the top 10 clones (P = 0.002) and lower entropy (P = 0.027). Gene expression analysis showed significant enrichment for upregulated negative regulation of immune system processes and downregulated T-cell activation and immune cell differentiation. Conclusion Our findings demonstrated that CD8-predominant AITL is a distinct immune pattern of AITL characterized by anti-tumor immunity impairment and an immunosuppressive microenvironment. These characteristics can interpret its severe clinical manifestations and poor outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuan Tang
- *Correspondence: Sha Zhao, ; Yuan Tang,
| | - Sha Zhao
- *Correspondence: Sha Zhao, ; Yuan Tang,
| |
Collapse
|
23
|
Zhang F, Li W, Cui Q, Chen Y, Liu Y. Case Report: Immune Microenvironment and Mutation Features in a Patient With Epstein–Barr Virus Positive Large B-Cell Lymphoma Secondary to Angioimmunoblastic T-Cell Lymphoma. Front Genet 2022; 13:940513. [PMID: 35938041 PMCID: PMC9354849 DOI: 10.3389/fgene.2022.940513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/20/2022] [Indexed: 01/24/2023] Open
Abstract
On rare occasions, secondary Epstein–Barr virus (EBV)-associated B-cell lymphoma can develop in patients with angioimmunoblastic T-cell lymphoma (AITL). Here, we describe the tumor microenvironment and mutation features of a patient with EBV + large B-cell lymphoma (LBCL) secondary to AITL. He was admitted to hospital due to a 1-year history of fever and enlarged right inguinal lymph nodes. A biopsy of the right inguinal lymph node demonstrated that numerous diffuse medium-sized atypical lymphocytes proliferated, together with increased extrafollicular follicular dendritic cell meshwork, and the lymphocytes expressed CD3, CD4, BCL6, CD10, PD-1, CXCL13, and Ki-67 (75%). Thus, a diagnosis of AITL was made. However, the disease progressed following treatment by CHOP regimen (cyclophosphamide, adriamycin, vincristine, and prednisone). Biopsy showed that most of the cells were positive for CD20 staining and IgH rearrangement. Analysis of 22 kinds of immune cells showed that the numbers of activated NK cells and activated memory T cells increased, while the T-follicular helper population decreased in the transformed sample. In addition, compared with the primary sample, RHOA (G17V) mutation was not detected, while JAK2 and TRIP12 gene mutations were detected in the transformed sample. Overall, we described the immune microenvironment and mutation features of a patient with EBV + LBCL secondary to AITL. This study will help us to understand the mechanisms by which AITL transforms to B-cell lymphoma.
Collapse
|
24
|
Attygalle AD, Dobson R, Chak PK, Vroobel KM, Wren D, Mugalaasi H, Morgan Y, Kaur M, Ahmad R, Chen Z, Naresh KN, Du M. Parallel evolution of two distinct lymphoid proliferations in clonal haematopoiesis. Histopathology 2022; 80:847-858. [PMID: 35064935 PMCID: PMC9310594 DOI: 10.1111/his.14619] [Citation(s) in RCA: 15] [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: 09/10/2021] [Revised: 01/07/2022] [Accepted: 01/14/2022] [Indexed: 11/28/2022]
Abstract
AIMS Angioimmunoblastic T-cell lymphoma (AITL) is genetically characterized by TET2 and DNMT3A mutations occurring in haematopoietic progenitor cells, and late events (e.g. the RHOA-G17V mutation) associated with malignant transformation. As TET2/DNMT3A-mutated progenitor cells can differentiate into multilineage progenies and give rise to both AITL and myeloid neoplasms, they may also have the potential to lead to other metachronous/synchronous neoplasms. We report two cases showing parallel evolution of two distinct potentially neoplastic lymphoid proliferations from a common mutated haematopoietic progenitor cell population. METHODS AND RESULTS Both cases presented with generalized lymphadenopathy. In case 1 (a 67-year-old female), an initial lymph node (LN) biopsy was dismissed as reactive, but a repeat biopsy showed a nodal marginal zone lymphoma (NMZL)-like proliferation with an increase in the number of T-follicular helper (TFH) cells. Immunohistochemistry, and clonality and mutational analyses by targeted sequencing of both whole tissue sections and microdissected NMZL-like lesions, demonstrated a clonal B-cell proliferation that harboured the BRAF-G469R mutation and shared TET2 and DNMT3A mutations with an underlying RHOA-G17V-mutant TFH proliferation. Review of the original LN biopsy showed histological and immunophenotypic features of AITL. In case 2 (a 66-year-old male), cytotoxic T-cell lymphoma with an increase in the number of Epstein-Barr virus-positive large B cells was diagnosed on initial biopsy. On review together with the relapsed biopsy, we identified an additional occult neoplastic TFH proliferation/smouldering AITL. Both T-cell proliferations shared TET2 and DNMT3A mutations while RHOA-G17V was confined to the smouldering AITL. CONCLUSIONS In addition to demonstrating diagnostic challenges, these cases expand the potential of clonal haematopoiesis in the development of different lineage neoplastic proliferations.
Collapse
Affiliation(s)
| | - Rachel Dobson
- Division of Cellular and Molecular Pathology, Department of PathologyUniversity of CambridgeCambridgeUK
| | - Pui Kwan Chak
- Department of Anatomical and Cellular PathologyPrince of Wales HospitalShatinNew TerritoriesHong Kong
| | | | - Dorte Wren
- Genomic Diagnostics Laboratory, Manchester Centre for Genomic MedicineManchester University NHS Foundation Trust, Saint Mary's HospitalManchesterUK
| | - Hood Mugalaasi
- Clinical Genomics, Haematological Diagnostic Malignancy ServiceThe Royal Marsden HospitalSuttonUK
| | - Yvonne Morgan
- Clinical Genomics, Haematological Diagnostic Malignancy ServiceThe Royal Marsden HospitalSuttonUK
| | - Manmit Kaur
- Department of HaematologyLuton & Dunstable University HospitalLutonUK
| | - Raida Ahmad
- Department of Cellular PathologyImperial College Healthcare NHS Trust, Charing Cross HospitalLondonUK
| | - Zi Chen
- Division of Cellular and Molecular Pathology, Department of PathologyUniversity of CambridgeCambridgeUK
| | - Kikkeri N Naresh
- Centre for HaematologyImperial College LondonLondonUK
- Clinical Research DivisionFred Hutchison Cancer Research CenterSeattleWAUSA
| | - Ming‐Qing Du
- Division of Cellular and Molecular Pathology, Department of PathologyUniversity of CambridgeCambridgeUK
| |
Collapse
|