1
|
Xing Y, Huang J, Zhang Y, Wang Y, Qi S. Advancing the understanding and management of angioimmunoblastic T-cell lymphoma: insights into its pathogenesis, clinical features, and emerging therapeutic strategies. Front Oncol 2025; 15:1479179. [PMID: 40098700 PMCID: PMC11911338 DOI: 10.3389/fonc.2025.1479179] [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: 08/11/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is a clinically aggressive non-Hodgkin lymphoma associated with many immune disorders. The incidence of AITL has gradually increased in Asia in recent years. Malignant AITL cells originate from T follicular helper cells, which have a unique morphology and complex biological characteristics. High-throughput sequencing studies have identified many gene mutations associated with its pathogenesis, including mutations in tet methylcytosine dioxygenase 2 (TET2), isocitrate dehydrogenase (NADP+) 2 (IDH2), DNA methyltransferase 3 alpha (DNMT3A), ras homolog family member A (RHOA), and T cell receptor-related genes. Currently, there is no standardized treatment for AITL, the first-line chemotherapy is ineffective, the recurrence rate is high, the overall prognosis of patients is poor, and the median survival time does not exceed three years. New drugs are urgently needed. However, with continuous in-depth study of the molecular genetic mechanism of AITL, some new drugs and therapies have been tested for patients with relapsed and refractory AITL, achieving some therapeutic effects. Increasing clinical studies are evaluating new potential targets for AITL based on specific molecular markers, gradually improving individualized treatment and ultimately improving the clinical prognosis of patients with AITL. This review first summarizes the progress of research on the etiology, clinical pathological characteristics, and molecular genetic mechanisms of AITL to enhance understanding of the disease. It then summarizes the progress of research on its treatment strategies to provide some references for clinically diagnosing and treating AITL.
Collapse
Affiliation(s)
- Yurou Xing
- Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junmeng Huang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Zhang
- Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongsheng Wang
- Thoracic Oncology Ward, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shaochong Qi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, China
| |
Collapse
|
2
|
Luo Q, Zhou L, Luo D, Yu L. Clonal hematopoiesis of indeterminate potential (CHIP): A potential contributor to lymphoma. Crit Rev Oncol Hematol 2025; 206:104589. [PMID: 39667716 DOI: 10.1016/j.critrevonc.2024.104589] [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: 08/13/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024] Open
Abstract
Clonal hematopoiesis (CH) typically refers to the clonal expansion of hematopoietic stem cells (HSCs) due to genetic mutations, serving as the pathogenic basis for various diseases. Clonal hematopoiesis of indeterminate potential (CHIP) is a subtype of CH, emerging as a significant risk factor for myeloid malignancies and cardiovascular diseases, which has attracted increasing attention. However, recent research has unveiled previously overlooked links between CHIP and lymphoma. This paper reviews the relationship between CHIP and lymphoma, focusing on the role and mechanism of TET2 and DNMT3A-mediated CHIP in lymphoma from the perspective of laboratory research and clinical observation. Additionally, we explore the therapeutic implications of targeting CHIP genes and inflammatory pathways in lymphoma. Our findings underscore the multifaceted influence of CHIP on lymphoma development and provide a promising avenue for therapeutic interventions in CHIP mediated lymphoma.
Collapse
Affiliation(s)
- QingQing Luo
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Provincial Key Laboratory of Hematological Diseases (2024SSY06052), Nanchang, Jiangxi, China
| | - LiLi Zhou
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Provincial Key Laboratory of Hematological Diseases (2024SSY06052), Nanchang, Jiangxi, China
| | - DaYa Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Li Yu
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Provincial Key Laboratory of Hematological Diseases (2024SSY06052), Nanchang, Jiangxi, China.
| |
Collapse
|
3
|
Chen F, Qiu S, Gui A, Jiang S, Yan Y, Wu J, Chen G, Zhu S, Liu Y, Xia Z, Yu B, Sun X, Gu JJ, Wang L, Liu W, Yang L, Zhang Q, Zuo J. Reduced OTUD7B expression correlates with poor prognosis in PTCL via non-canonical NF-κB. Int J Hematol 2025; 121:194-205. [PMID: 39636331 DOI: 10.1007/s12185-024-03877-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Peripheral T cell lymphoma (PTCL) is an aggressive and highly heterogeneous lymphoma with a bleak prognosis, highlighting the urgent need for an effective biomarker to guide therapeutic strategies. Ovarian tumor domain-containing 7B (OTUD7B) has been shown to have a critical function in the progression of cancers. However, the prognostic significance of OTUD7B in PTCL remains unexplored. In this study, we demonstrated for the first time that PTCL patients with low expression of OTUD7B had shorter progression-free survival (PFS) and overall survival (OS). In addition, OTUD7B knockdown promoted chemoresistance to doxorubicin in PTCL cell lines, and led to increased translocation of p52 from the cytoplasm to the nucleus. Inhibition of non-canonical NF-κB partially restored the sensitivity of PTCL cells to doxorubicin. Remarkably, 5-azacytidine and cytarabine upregulated the expression of OTUD7B and exhibited a synergistic anti-lymphoma effect in PTCL. In summary, our study confirmed the prognostic role of OTUD7B in PTCL and the promising therapeutic potential of combining 5-azacytidine or cytarabine and doxorubicin for PTCL treatment.
Collapse
MESH Headings
- Humans
- Prognosis
- NF-kappa B/metabolism
- Doxorubicin/pharmacology
- Female
- Cell Line, Tumor
- Lymphoma, T-Cell, Peripheral/genetics
- Lymphoma, T-Cell, Peripheral/mortality
- Lymphoma, T-Cell, Peripheral/drug therapy
- Lymphoma, T-Cell, Peripheral/metabolism
- Lymphoma, T-Cell, Peripheral/diagnosis
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic
- Middle Aged
- Male
Collapse
Affiliation(s)
- Feng Chen
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Shi Qiu
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Ailing Gui
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Shiyu Jiang
- Department of Lymphoma, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yichen Yan
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jichuan Wu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Guangliang Chen
- Department of Lymphoma, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Shun Zhu
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yizhen Liu
- Department of Lymphoma, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Zuguang Xia
- Department of Lymphoma, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Baohua Yu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Xiaojian Sun
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Jennifer Gu
- Department of Medicine & Immunology, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Lan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wen Liu
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Ling Yang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Qunling Zhang
- Department of Lymphoma, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Ji Zuo
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
4
|
Xie W, Li X, Chen H, Chu J, Zhang L, Tang B, Huang W, Li L, Lin J, Dong Y. 5-Hydroxymethylcytosine Profiles of cfDNA in Urine as Diagnostic, Differential Diagnosis and Prognostic Markers for Multiple Myeloma. Cancer Med 2024; 13:e70477. [PMID: 39711442 DOI: 10.1002/cam4.70477] [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: 05/17/2024] [Revised: 11/08/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND An effective urine-based method for the diagnosis, differential diagnosis and prognosis of multiple myeloma (MM) has not yet been developed. Urine cell-free DNA (cfDNA) carrying cancer-specific genetic and epigenetic aberrations may enable a noninvasive "liquid biopsy" for diagnosis and monitoring of cancer. METHODS We first identified MM-specific hydroxymethylcytosine signatures by comparing 64 MM patients, 23 amyloidosis (AM) patients and 59 healthy cohort. Then, we applied a machine learning algorithm to develop diagnostic and differential diagnosis model. Finally, the prognosis of MM patients was predicted based on their survival time at the last follow-up. RESULTS We identified 11 5hmC markers using logistic regression algorithm could effectively diagnosis MM (AUC = 0.902), and achieved 85.00% specificity and 85.71% sensitivity. These 11 markers could also effectively differential diagnosis MM (AUC = 0.805) with 88.89% specificity and 73.08% sensitivity. In addition, the prognostic prediction model also effectively predicted the prognosis of patients with MM (p < 0.01), of which 4 differential markers (RAPGEF2, BRD1, TET2, TRAF3IP2) could independently predict the prognosis of MM. CONCLUSIONS Together, our findings showed the value of urine cfDNA hydroxymethylcytosine markers in the diagnosis, differential diagnosis and prognosis of MM. Meantime, our study provides a promising and completely non-invasive method for the diagnosis, differential diagnosis and prognosis prediction of MM.
Collapse
Affiliation(s)
- Weiwei Xie
- Department of Hematology, Peking University First Hospital, Beijing, People's Republic of China
| | - Xuehui Li
- Department of Pharmacology, Xinjiang Medical University, Urumqi, China
| | - Hangyu Chen
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
- Synthetic and Functional Biomolecules Center, Peking University, Beijing, China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- Peking University Third Hospital Cancer Center, Beijing, China
| | - Jinlin Chu
- Department of Pharmacology, Xinjiang Medical University, Urumqi, China
| | - Lei Zhang
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
- Synthetic and Functional Biomolecules Center, Peking University, Beijing, China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- Peking University Third Hospital Cancer Center, Beijing, China
| | - Bo Tang
- Department of Hematology, Peking University First Hospital, Beijing, People's Republic of China
| | - Wenrong Huang
- Department of Hematology, Fifth Medical Center, General Hospital of the People's Liberation Army, Beijing, People's Republic of China
| | - Linlin Li
- Department of Pharmacology, Xinjiang Medical University, Urumqi, China
- Key Laboratory of Active Components of Xinjiang Natural Medicine and Drug Release Technology, Urumqi, China
| | - Jian Lin
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
- Synthetic and Functional Biomolecules Center, Peking University, Beijing, China
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- Peking University Third Hospital Cancer Center, Beijing, China
| | - Yujun Dong
- Department of Hematology, Peking University First Hospital, Beijing, People's Republic of China
| |
Collapse
|
5
|
Azimpouran M, Bui CM, Balzer B, Kitahara S. Rapidly Progressive Primary Cutaneous Gamma Delta T-Cell Lymphoma With FYN Gene Alteration. Am J Dermatopathol 2024; 46:e120-e123. [PMID: 39412302 DOI: 10.1097/dad.0000000000002856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
ABSTRACT Primary cutaneous gamma delta T-cell lymphoma (PCGDTCL) is a rare type of non-Hodgkin lymphoma accounting for <1% of primary cutaneous T-cell lymphomas. The exact cause of PCGDTCL is not known, however, it is thought that chronic antigen exposure in the skin may lead to immune dysregulation at the site, resulting in abnormal proliferation of mature, post-thymic cytotoxic gamma delta T cells. Mutations are the most common genetic alteration seen in PCGDTCL, while structural abnormalities such as gene fusions are not common. We report a case of PCGDTCL with atypical immunophenotypic features, including expression of CD5 with lack of cytotoxic marker expression, and a structural alteration leading to FYN deletion at exon 8. Recently, it was described that a deletion of the area between FYN exon 8 and TRAF3IP2 intron 2 results in a novel FYN::TRAF3IP2 fusion in peripheral T-cell lymphoma, not otherwise specified. We describe our patient's clinical course, differential diagnosis, and potential implications of FYN deletion on disease pathogenesis. To our knowledge, this is the first report of an FYN structural alteration to be described in PCGDTCL.
Collapse
Affiliation(s)
- Mahzad Azimpouran
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | | | | |
Collapse
|
6
|
Iqbal J, Inghirami G, Chan WC. New insights into the biology of T-cell lymphomas. Blood 2024; 144:1873-1886. [PMID: 39213420 PMCID: PMC11551850 DOI: 10.1182/blood.2023021787] [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: 01/08/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
ABSTRACT Peripheral T-cell lymphomas (PTCLs) encompass a heterogeneous group of postthymic T-cell lymphomas with >30 distinct subtypes associated with varied clinicopathological features. Unfortunately, the overall survival of the major PTCL subtypes is dismal and has not improved for decades; thus, there is an urgent unmet clinical need to improve diagnosis, therapies, and clinical outcomes. The diagnosis is often challenging, requiring a combinatorial evaluation of clinical, morphologic, and immunophenotypic features. PTCL pathobiology is difficult to investigate due to enormous intertumor and intratumor heterogeneity, limited tissue availability, and the paucity of authentic T-cell lymphoma cell lines or genetically faithful animal models. The application of transcriptomic profiling and genomic sequencing has markedly accelerated the discovery of new biomarkers, molecular signatures, and genetic lesions, and some of the discoveries have been included in the revised World Health Organization or International Consensus Classification. Genome-wide investigations have revealed the mutational landscape and transcriptomic profiles of PTCL entities, defined the cell of origin as a major determinant of T-cell lymphoma biology, and allowed for the refinement of biologically and clinically meaningful entities for precision therapy. In this review, we prioritize the discussion on common nodal PTCL subtypes together with 2 virus-associated T-cell and natural killer cell lymphomas. We succinctly review normal T-cell development, differentiation, and T-cell receptor signaling as they relate to PTCL pathogenesis and biology. This review will facilitate a better biological understanding of the different PTCL entities and their stratification for additional studies and target-directed clinical trials.
Collapse
Affiliation(s)
- Javeed Iqbal
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE
| | - Giorgio Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Wing C. Chan
- Department of Pathology, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
7
|
Liu F, Zhang J, Gu X, Guo Q, Guo W. Single-cell transcriptome sequencing reveals SPP1-CD44-mediated macrophage-tumor cell interactions drive chemoresistance in TNBC. J Cell Mol Med 2024; 28:e18525. [PMID: 38982317 PMCID: PMC11233263 DOI: 10.1111/jcmm.18525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/28/2024] [Accepted: 06/12/2024] [Indexed: 07/11/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is often considered one of the most aggressive subtypes of breast cancer, characterized by a high recurrence rate and low overall survival (OS). It is notorious for posing challenges related to drug resistance. While there has been progress in TNBC research, the mechanisms underlying chemotherapy resistance in TNBC remain largely elusive. We collect single-cell RNA sequencing (scRNA-seq) data from five TNBC patients susceptible to chemotherapy and five resistant cases. Comprehensive analyses involving copy number variation (CNV), pseudotime trajectory, cell-cell interactions, pseudospace analysis, as well as transcription factor and functional enrichment are conducted specifically on macrophages and malignant cells. Furthermore, we performed validation experiments on clinical samples using multiplex immunofluorescence. We identified a subset of SPP1+ macrophages that secrete SPP1 signals interacting with CD44 on malignant cell surfaces, potentially activating the PDE3B pathway within malignant cells via the integrin pathway, leading to chemotherapy resistance. The abnormally enhanced SPP1 signal between macrophages and malignant cells may serve as a factor promoting chemotherapy resistance in TNBC patients. Therefore, SPP1+ macrophages could potentially serve as a therapeutic target to reduce chemotherapy resistance.
Collapse
Affiliation(s)
- Fuzhong Liu
- Xinjiang Medical University Affiliated Cancer HospitalUrumqiChina
| | - Junfeng Zhang
- Xinjiang Medical University Affiliated Cancer HospitalUrumqiChina
| | - Xiaowei Gu
- Xinjiang Medical University Affiliated Cancer HospitalUrumqiChina
| | | | - Wenjia Guo
- Xinjiang Medical University Affiliated Cancer HospitalUrumqiChina
- Xinjiang Key Laboratory of Translational Biomedical EngineeringUrumqiChina
| |
Collapse
|
8
|
Chang EWY, Tan YH, Chan JY. Novel clinical risk stratification and treatment strategies in relapsed/refractory peripheral T-cell lymphoma. J Hematol Oncol 2024; 17:38. [PMID: 38824603 PMCID: PMC11144347 DOI: 10.1186/s13045-024-01560-7] [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: 03/18/2024] [Accepted: 05/26/2024] [Indexed: 06/03/2024] Open
Abstract
Peripheral T cell lymphoma (PTCL) represents a group of heterogeneous hematological malignancies, which are notoriously challenging to treat and outcomes are typically poor. Over the past two decades, clinical prognostic indices for patient risk stratification have evolved, while several targeted agents are now available to complement combination chemotherapy in the frontline setting or as a salvage strategy. With further understanding of the molecular pathobiology of PTCL, several innovative approaches incorporating immunomodulatory agents, epigenetic therapies, oncogenic kinase inhibitors and immunotherapeutics have come to the forefront. In this review, we provide a comprehensive overview of the progress in developing clinical prognostic indices for PTCL and describe the broad therapeutic landscape, emphasizing novel targetable pathways that have entered early phase clinical studies.
Collapse
Affiliation(s)
- Esther Wei Yin Chang
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.
- Duke-NUS Medical School, Singapore, Singapore.
| | - Ya Hwee Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Jason Yongsheng Chan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore.
- Duke-NUS Medical School, Singapore, Singapore.
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore, Singapore.
| |
Collapse
|
9
|
Bisig B, Savage KJ, De Leval L. Pathobiology of nodal peripheral T-cell lymphomas: current understanding and future directions. Haematologica 2023; 108:3227-3243. [PMID: 38037800 PMCID: PMC10690915 DOI: 10.3324/haematol.2023.282716] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 08/23/2023] [Indexed: 12/02/2023] Open
Abstract
Predominantly nodal is the most common clinical presentation of peripheral T- (and NK-) cell lymphomas (PTCL), which comprise three main groups of diseases: (i) systemic anaplastic large cell lymphomas (ALCL), whether positive or negative for anaplastic lymphoma kinase (ALK); (ii) follicular helper T-cell lymphomas (TFHL); and (iii) PTCL, not otherwise specified (NOS). Recent advances in the genomic and molecular characterization of PTCL, with enhanced understanding of pathobiology, have translated into significant updates in the latest 2022 classifications of lymphomas. ALK-negative ALCL is now recognized to be genetically heterogeneous, with identification of DUSP22 rearrangements in approximately 20-30% of cases, correlated with distinctive pathological and biological features. The notion of cell-of-origin as an important determinant of the classification of nodal PTCL is best exemplified by TFHL, considered as one disease or a group of related entities, sharing oncogenic pathways with frequent recurrent epigenetic mutations as well as a relationship to clonal hematopoiesis. Data are emerging to support that a similar cell-of-origin concept might be relevant to characterize meaningful subgroups within PTCL, NOS, based on cytotoxic and/or Th1 versus Th2 signatures. The small group of primary nodal Epstein-Barr virus-positive lymphomas of T- or NK-cell derivation, formerly considered PTCL, NOS, is now classified separately, due to distinctive features, and notably an aggressive course. This review summarizes current knowledge of the pathology and biology of nodal-based PTCL entities, with an emphasis on recent findings and underlying oncogenic mechanisms.
Collapse
Affiliation(s)
- Bettina Bisig
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne
| | - Kerry J Savage
- Centre for Lymphoid Cancer, Division of Medical Oncology, BC Cancer and University of British Columbia, Vancouver, British Columbia
| | - Laurence De Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne.
| |
Collapse
|
10
|
Hu D, Mo X, Jihang L, Huang C, Xie H, Jin L. Novel diagnostic biomarkers of oxidative stress, immunological characterization and experimental validation in Alzheimer's disease. Aging (Albany NY) 2023; 15:10389-10406. [PMID: 37801482 PMCID: PMC10599743 DOI: 10.18632/aging.205084] [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: 05/21/2023] [Accepted: 09/02/2023] [Indexed: 10/08/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition causing cognitive decline. Oxidative stress (OS) is believed to contribute to neuronal death and dysfunction in AD. We conducted a study to identify differentially expressed OS-related genes (DEOSGs) through bioinformatics analysis and experimental validation, aiming to develop a diagnostic model for AD. We analyzed the GSE33000 dataset to identify OS regulator expression profiles and create molecular clusters (C1 and C2) associated with immune cell infiltration using 310 AD samples. Cluster analysis revealed significant heterogeneity in immune infiltration. The 'WGCNA' algorithm identified cluster-specific and disease-specific differentially expressed genes (DGEs). Four machine learning models (random forest (RF), support vector machine (SVM), generalized linear model (GLM) and extreme gradient boosting (XGB)) were compared, with GLM performing the best (AUC = 0.812). Five DEOSGs (NFKBIA, PLCE1, CLIC1, SLCO4A1, TRAF3IP2) were identified based on the GLM model. AD subtype prediction accuracy was validated using nomograms and calibration curves. External datasets (GSE122063 and GSE106241) confirmed the expression levels and clinical significance of important genes. Experimental validation through RT-qPCR showed increased expression of NFKBIA, CLIC1, SLCO4A1, TRAF3IP2, and decreased expression of PLCE1 in the temporal cortex of AD mice. This study provides insights for AD research and treatment, particularly focusing on the five model-related DEOSGs.
Collapse
Affiliation(s)
- Di Hu
- Department of Neurology and Stroke Centre, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaocong Mo
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Luo Jihang
- Department of Oncology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- Department of Infectious Diseases, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Cheng Huang
- Department of Neurology and Stroke Centre, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hesong Xie
- Department of Neurology and Stroke Centre, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ling Jin
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| |
Collapse
|
11
|
Liu C, Li S, Tang Y. Advances in the expression and function of Fyn in different human tumors. Clin Transl Oncol 2023; 25:2852-2860. [PMID: 37093456 DOI: 10.1007/s12094-023-03167-9] [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: 11/13/2022] [Accepted: 03/18/2023] [Indexed: 04/25/2023]
Abstract
The tyrosine kinase Fyn is a member of the SRC family of kinases, and its sustained activation is closely linked to tumor cell migration, proliferation, and cell metabolism. Recently, Fyn has been found to be expressed in various tumor tissues, and the expression and function of Fyn vary between tumors, with Fyn acting as an oncogene to promote proliferation and metastasis in some tumors. This article summarizes the recent studies on the role of Fyn in different human tumors, focusing on the role of Fyn in melanoma, breast cancer, glioma, lung cancer, and peripheral T-cell lymphoma in order to provide a basis for future research and targeted therapy in different human tumors.
Collapse
Affiliation(s)
- Changqing Liu
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hunan Province, 28 Changsheng Road, Hengyang, 421001, People's Republic of China
| | - Shan Li
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hunan Province, 28 Changsheng Road, Hengyang, 421001, People's Republic of China
| | - Yunlian Tang
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hunan Province, 28 Changsheng Road, Hengyang, 421001, People's Republic of China.
| |
Collapse
|
12
|
Carty SA, Murga-Zamalloa CA, Wilcox RA. SOHO State of the Art Updates and Next Questions | New Pathways and New Targets in PTCL: Staying on Target. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:561-574. [PMID: 37142534 PMCID: PMC10565700 DOI: 10.1016/j.clml.2023.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/05/2023] [Accepted: 04/16/2023] [Indexed: 05/06/2023]
Abstract
While the peripheral T-cell lymphomas (PTCL) remain a therapeutic challenge, and increasingly account for a disproportionate number of lymphoma-related deaths, improved understanding of disease pathogenesis and classification, and the development of novel therapeutic agents over the past decade, all provide reasons for a more optimistic outlook in the next. Despite their genetic and molecular heterogeneity, many PTCL are dependent upon signaling input provided by antigen, costimulatory, and cytokine receptors. While gain-of-function alterations effecting these pathways are recurrently observed in many PTCL, more often than not, signaling remains ligand-and tumor microenvironment (TME)-dependent. Consequently, the TME and its constituents are increasingly recognized as "on target". Utilizing a "3 signal" model, we will review new-and old-therapeutic targets that are relevant for the more common nodal PTCL subtypes.
Collapse
Affiliation(s)
- Shannon A Carty
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI
| | | | - Ryan A Wilcox
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI.
| |
Collapse
|
13
|
Du J, Jin S, Zhang M, Fu X, Yang J, Zhang L, Chen Z, Huang Z, Li W, Hou J, Wang T. Precise diagnosis and targeted therapy of nodal T-follicular helper cell lymphoma (T-FHCL). Front Oncol 2023; 13:1163190. [PMID: 37188182 PMCID: PMC10175683 DOI: 10.3389/fonc.2023.1163190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Nodal T-follicular helper cell lymphoma (T-FHCL) derived from T-follicular helper (Tfh) cell falls into a heterogeneous category of peripheral T-cell lymphoma (PTCL). Due to the limited number of therapeutic regimens and limited first-line efficacy, T-FHCL has a poor prognosis, and there is an urgent need for effective targeted therapies. With advancements in sequencing technologies, especially single-cell sequencing and next-generation sequencing, more specific genetic aberrations characteristic of T-FHCL can be discovered, allowing for precise molecular diagnosis and specific research on novel agents. Many biomarker-targeting agents, used either alone or in combination, have been tested, and they have generally enhanced the therapeutic outcomes of T-FHCL. Histone deacetylase inhibitors achieve significant clinical benefits in the treatment of T-FHCL, especially in combination therapy. Chimeric antigen receptor T-cell (CAR-T-cell) immunotherapies, hematopoietic stem cell transplantation, and other potential agents merit further study.
Collapse
Affiliation(s)
- Jun Du
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shikai Jin
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minghui Zhang
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuehang Fu
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingwen Yang
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liwen Zhang
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenwei Chen
- Department of Clinical Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zoufang Huang
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Weisong Li
- Department of Pathology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jian Hou
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Wang
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
14
|
Li S, Liu C, Tang Y. Role of Fyn in hematological malignancies. J Cancer Res Clin Oncol 2023:10.1007/s00432-023-04608-2. [PMID: 36754870 DOI: 10.1007/s00432-023-04608-2] [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: 12/02/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND Tyrosine kinase Fyn is a member of the Src family of kinases. In addition to the wild type, three mRNA splice isoforms of Fyn have been identified; Fyn-B, Fyn-T, and Fyn-C. Fyn-T is highly expressed in T lymphocytes, and its expression level is significantly higher in mature T cells than in immature T cells. The abnormal expression of Fyn is closely related to the metabolism, proliferation, and migration of tumor cells. Recent studies have shown that Fyn is expressed in a variety of tumor tissues, and its expression and function vary among different tumors. In some tumors, Fyn acts as a pro-oncogene to promote tumor proliferation and metastasis. Moreover, Fyn mutations have been detected in many hematological tumors in recent years, suggesting a critical regulatory role of Fyn in the development of malignancies. METHODS This review analyzed the relevant literature in PubMed and other databases. PURPOSE The aim of this study was to systemically review recent research findings on various aspects of Fyn in the pathogenesis and treatment of different types of hematological malignancies and suggests possible future research directions for targeted tumor therapy. CONCLUSION Fyn could be a novel prognostic marker and therapeutic target. Treatment option targeting Fyn might be beneficial for future studies.
Collapse
Affiliation(s)
- Shan Li
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Changqing Liu
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yunlian Tang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
15
|
Piccaluga PP, Cascianelli C, Inghirami G. Tyrosine kinases in nodal peripheral T-cell lymphomas. Front Oncol 2023; 13:1099943. [PMID: 36845713 PMCID: PMC9946040 DOI: 10.3389/fonc.2023.1099943] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
Nodal peripheral T-cell lymphomas (PTCL) are uncommon and heterogeneous tumors characterized by a dismal prognosis. Targeted therapy has been proposed. However, reliable targets are mostly represented by a few surface antigens (e.g., CD52 and CD30), chemokine receptors (e.g., CCR4), and epigenetic gene expression regulation. In the last two decades, however, several studies have supported the idea that tyrosine kinase (TK) deregulation might be relevant for both the pathogenesis and treatment of PTCL. Indeed, they can be expressed or activated as a consequence of their involvement in genetic lesions, such as translocations, or by ligand overexpression. The most striking example is ALK in anaplastic large-cell lymphomas (ALCL). ALK activity is necessary to support cell proliferation and survival, and its inhibition leads to cell death. Notably, STAT3 was found to be the main downstream ALK effector. Other TKs are consistently expressed and active in PTCLs, such as PDGFRA, and members of the T-cell receptor signaling family, such as SYK. Notably, as in the case of ALK, STAT proteins have emerged as key downstream factors for most of the involved TK.
Collapse
Affiliation(s)
- Pier Paolo Piccaluga
- Biobank of Research, IRCCS Azienda Opedaliera-Universitaria di Bologna, Bologna, Italy
- Department of Experimental, Diagnostic, and Specialty Medicine, School of Medicine, University of Bologna, Bologna, Italy
| | - Chiara Cascianelli
- Biobank of Research, IRCCS Azienda Opedaliera-Universitaria di Bologna, Bologna, Italy
| | - Giorgio Inghirami
- Immunopathology and Hematopathology, Weill Cornell Medical College, New York-Presbyterian Hospital, New York, NY, United States
| |
Collapse
|
16
|
Peng S, Fu Y. FYN: emerging biological roles and potential therapeutic targets in cancer. J Transl Med 2023; 21:84. [PMID: 36740671 PMCID: PMC9901160 DOI: 10.1186/s12967-023-03930-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/25/2023] [Indexed: 02/07/2023] Open
Abstract
Src family protein kinases (SFKs) play a key role in cell adhesion, invasion, proliferation, survival, apoptosis, and angiogenesis during tumor development. In humans, SFKs consists of eight family members with similar structure and function. There is a high level of overexpression or hyperactivity of SFKs in tumor, and they play an important role in multiple signaling pathways involved in tumorigenesis. FYN is a member of the SFKs that regulate normal cellular processes. Additionally, FYN is highly expressed in many cancers and promotes cancer growth and metastasis through diverse biological functions such as cell growth, apoptosis, and motility migration, as well as the development of drug resistance in many tumors. Moreover, FYN is involved in the regulation of multiple cancer-related signaling pathways, including interactions with ERK, COX-2, STAT5, MET and AKT. FYN is therefore an attractive therapeutic target for various tumor types, and suppressing FYN can improve the prognosis and prolong the life of patients. The purpose of this review is to provide an overview of FYN's structure, expression, upstream regulators, downstream substrate molecules, and biological functions in tumors.
Collapse
Affiliation(s)
- SanFei Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Yang Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| |
Collapse
|
17
|
Mutations Affecting Genes in the Proximal T-Cell Receptor Signaling Pathway in Peripheral T-Cell Lymphoma. Cancers (Basel) 2022; 14:cancers14153716. [PMID: 35954378 PMCID: PMC9367541 DOI: 10.3390/cancers14153716] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary The advent of next-generation sequencing (NGS) has allowed rapid advances in genomic studies on the pathogenesis and biology of peripheral T-cell lymphoma (PTCL). Recurrent mutations and fusions in genes related to the proximal TCR signaling pathway have been identified and show an important pathogenic role in PTCL. In this review, we summarize the genomic alterations in TCR signaling identified in different subgroups of PTCL patients and the functional impact of these alterations on TCR signaling and downstream pathways. We also discuss novel agents that could target TCR-related mutations and may hold promise for improving the treatment of PTCL. Abstract Peripheral T-cell lymphoma (PTCL) comprises a heterogeneous group of mature T-cell malignancies. Recurrent activating mutations and fusions in genes related to the proximal TCR signaling pathway have been identified in preclinical and clinical studies. This review summarizes the genetic alterations affecting proximal TCR signaling identified from different subgroups of PTCL and the functional impact on TCR signaling and downstream pathways. These genetic abnormalities include mostly missense mutations, occasional indels, and gene fusions involving CD28, CARD11, the GTPase RHOA, the guanine nucleotide exchange factor VAV1, and kinases including FYN, ITK, PLCG1, PKCB, and PI3K subunits. Most of these aberrations are activating mutations that can potentially be targeted by inhibitors, some of which are being tested in clinical trials that are briefly outlined in this review. Finally, we focus on the molecular pathology of recently identified subgroups of PTCL-NOS and highlight the unique genetic profiles associated with PTCL-GATA3.
Collapse
|
18
|
Cortes JR, Filip I, Albero R, Patiño-Galindo JA, Quinn SA, Lin WHW, Laurent AP, Shih BB, Brown JA, Cooke AJ, Mackey A, Einson J, Zairis S, Rivas-Delgado A, Laginestra MA, Pileri S, Campo E, Bhagat G, Ferrando AA, Rabadan R, Palomero T. Oncogenic Vav1-Myo1f induces therapeutically targetable macrophage-rich tumor microenvironment in peripheral T cell lymphoma. Cell Rep 2022; 39:110695. [PMID: 35443168 PMCID: PMC9059228 DOI: 10.1016/j.celrep.2022.110695] [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: 07/14/2021] [Revised: 01/26/2022] [Accepted: 03/25/2022] [Indexed: 11/03/2022] Open
Abstract
Peripheral T cell lymphoma not otherwise specified (PTCL-NOS) comprises heterogeneous lymphoid malignancies characterized by pleomorphic lymphocytes and variable inflammatory cell-rich tumor microenvironment. Genetic drivers in PTCL-NOS include genomic alterations affecting the VAV1 oncogene; however, their specific role and mechanisms in PTCL-NOS remain incompletely understood. Here we show that expression of Vav1-Myo1f, a recurrent PTCL-associated VAV1 fusion, induces oncogenic transformation of CD4+ T cells. Notably, mouse Vav1-Myo1f lymphomas show T helper type 2 features analogous to high-risk GATA3+ human PTCL. Single-cell transcriptome analysis reveals that Vav1-Myo1f alters T cell differentiation and leads to accumulation of tumor-associated macrophages (TAMs) in the tumor microenvironment, a feature linked with aggressiveness in human PTCL. Importantly, therapeutic targeting of TAMs induces strong anti-lymphoma effects, highlighting the lymphoma cells' dependency on the microenvironment. These results demonstrate an oncogenic role for Vav1-Myo1f in the pathogenesis of PTCL, involving deregulation in T cell polarization, and identify the lymphoma-associated macrophage-tumor microenvironment as a therapeutic target in PTCL.
Collapse
Affiliation(s)
- Jose R Cortes
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Ioan Filip
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Robert Albero
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | | | - S Aidan Quinn
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Wen-Hsuan W Lin
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Anouchka P Laurent
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Bobby B Shih
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Jessie A Brown
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Anisha J Cooke
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Adam Mackey
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA
| | - Jonah Einson
- Department of Biomedical Informatics, Columbia University, New York, NY 10032, USA
| | - Sakellarios Zairis
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | | | | | - Stefano Pileri
- Division of Hematopathology, European Institute of Oncology IRCCS, Milan 20141, Italy
| | - Elias Campo
- Hematopathology Unit, Department of Pathology, Hospital Clínic-IDIBAPS, Barcelona 08036, Spain
| | - Govind Bhagat
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Adolfo A Ferrando
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA; Department of Systems Biology, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Raul Rabadan
- Department of Systems Biology, Columbia University, New York, NY 10032, USA; Department of Biomedical Informatics, Columbia University, New York, NY 10032, USA
| | - Teresa Palomero
- Institute for Cancer Genetics, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
19
|
USP8 inhibition reshapes an inflamed tumor microenvironment that potentiates the immunotherapy. Nat Commun 2022; 13:1700. [PMID: 35361799 PMCID: PMC8971425 DOI: 10.1038/s41467-022-29401-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 03/15/2022] [Indexed: 01/20/2023] Open
Abstract
Anti-PD-1/PD-L1 immunotherapy has achieved impressive therapeutic outcomes in patients with multiple cancer types. However, the underlined molecular mechanism(s) for moderate response rate (15–25%) or resistance to PD-1/PD-L1 blockade remains not completely understood. Here, we report that inhibiting the deubiquitinase, USP8, significantly enhances the efficacy of anti-PD-1/PD-L1 immunotherapy through reshaping an inflamed tumor microenvironment (TME). Mechanistically, USP8 inhibition increases PD-L1 protein abundance through elevating the TRAF6-mediated K63-linked ubiquitination of PD-L1 to antagonize K48-linked ubiquitination and degradation of PD-L1. In addition, USP8 inhibition also triggers innate immune response and MHC-I expression largely through activating the NF-κB signaling. Based on these mechanisms, USP8 inhibitor combination with PD-1/PD-L1 blockade significantly activates the infiltrated CD8+ T cells to suppress tumor growth and improves the survival benefit in several murine tumor models. Thus, our study reveals a potential combined therapeutic strategy to utilize a USP8 inhibitor and PD-1/PD-L1 blockade for enhancing anti-tumor efficacy. The regulatory mechanisms of PD-L1 posttranslational modifications are not completely understood. Here the authors show that USP8 negatively regulates PD-L1 protein abundance by removing the K63-linked ubiquitination of PD-L1; while USP8 inhibition increases MHC-I expression and triggers anti-tumour immune responses through activating NF-κB signalling.
Collapse
|
20
|
Yu DD, Zhang J. Update on recurrent mutations in angioimmunoblastic T-cell lymphoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2021; 14:1108-1118. [PMID: 35027991 PMCID: PMC8748014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/26/2021] [Indexed: 06/14/2023]
Abstract
Angioimmunoblastic T-cell lymphoma (AITL) is a subtype of peripheral T cell lymphoma (PTCL), defined by genetic alterations that induce abnormal immune activity and inflammatory disorders. Through recent discoveries using genomic studies, the identification of various recurrent mutations has provided greater insight and changed our understanding of the molecular genetics of the disease. By acknowledging these recurrent mutations and their affected pathways, the diagnosis, prognosis, treatment, and survival of AITL can be improved. In this review, we summarize the known recurrent mutations present in the molecular pathogenesis of AITL by emphasizing the effects of mutations on signaling pathways and genes, as well as the multistep process of AITL development.
Collapse
Affiliation(s)
| | - Jianzhong Zhang
- Department of Pathology, Strategic Support Force Medical CenterBeijing, China
| |
Collapse
|
21
|
Robles-Valero J, Fernández-Nevado L, Lorenzo-Martín LF, Cuadrado M, Fernández-Pisonero I, Rodríguez-Fdez S, Astorga-Simón EN, Abad A, Caloto R, Bustelo XR. Cancer-associated mutations in VAV1 trigger variegated signaling outputs and T-cell lymphomagenesis. EMBO J 2021; 40:e108125. [PMID: 34617326 PMCID: PMC8591544 DOI: 10.15252/embj.2021108125] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 09/04/2021] [Accepted: 09/09/2021] [Indexed: 12/25/2022] Open
Abstract
Mutations in VAV1, a gene that encodes a multifunctional protein important for lymphocytes, are found at different frequencies in peripheral T‐cell lymphoma (PTCL), non‐small cell lung cancer, and other tumors. However, their pathobiological significance remains unsettled. After cataloguing 51 cancer‐associated VAV1 mutations, we show here that they can be classified in five subtypes according to functional impact on the three main VAV1 signaling branches, GEF‐dependent activation of RAC1, GEF‐independent adaptor‐like, and tumor suppressor functions. These mutations target new and previously established regulatory layers of the protein, leading to quantitative and qualitative changes in VAV1 signaling output. We also demonstrate that the most frequent VAV1 mutant subtype drives PTCL formation in mice. This process requires the concurrent engagement of two downstream signaling branches that promote the chronic activation and transformation of follicular helper T cells. Collectively, these data reveal the genetic constraints associated with the lymphomagenic potential of VAV1 mutant subsets, similarities with other PTCL driver genes, and potential therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Javier Robles-Valero
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Lucía Fernández-Nevado
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - L Francisco Lorenzo-Martín
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Myriam Cuadrado
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Isabel Fernández-Pisonero
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Sonia Rodríguez-Fdez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Elsa N Astorga-Simón
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain
| | - Antonio Abad
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Rubén Caloto
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| | - Xosé R Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, Salamanca, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, Salamanca, Spain
| |
Collapse
|
22
|
Qiu L, Cho JH, Jelloul FZ, Vega F. SOHO State of the Art Updates and Next Questions: Pathology and Pathogenesis of Nodal Peripheral T-Cell Lymphomas. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:287-296. [PMID: 34776400 DOI: 10.1016/j.clml.2021.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/30/2021] [Accepted: 10/09/2021] [Indexed: 10/20/2022]
Abstract
Peripheral T-cell lymphomas (PTCLs) are a heterogeneous and often clinically aggressive group of neoplasms derived from mature post-thymic T-lymphocytes. These neoplasms are rare and usually diagnostically challenging. Our understanding of the pathogenesis of PTCL is increasing and this improved knowledge is leading us to better molecular characterization, more objective and accurate diagnostic criteria, more effective risk assessment, and potentially better treatments. The focus of this paper is to present a brief overview of the current pathology criteria and molecular and genetic features of nodal peripheral T-cell lymphomas focusing on distinct genetically and molecularly defined subgroups that are being recognized within each major nodal PTCL category. It is expected that the molecular stratification will improve the diagnosis and will provide novel therapeutic opportunities (biomarker-driven and targeted therapies) that might benefit and change the outcomes of patients with these neoplasms.
Collapse
Affiliation(s)
- Lianqun Qiu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeong Hee Cho
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fatima Zahra Jelloul
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Francisco Vega
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas; UT Health Graduate School of Biomedical Sciences, Houston, Texas.
| |
Collapse
|
23
|
Debackere K, Marcelis L, Demeyer S, Vanden Bempt M, Mentens N, Gielen O, Jacobs K, Broux M, Verhoef G, Michaux L, Graux C, Wlodarska I, Gaulard P, de Leval L, Tousseyn T, Cools J, Dierickx D. Fusion transcripts FYN-TRAF3IP2 and KHDRBS1-LCK hijack T cell receptor signaling in peripheral T-cell lymphoma, not otherwise specified. Nat Commun 2021; 12:3705. [PMID: 34140493 PMCID: PMC8211700 DOI: 10.1038/s41467-021-24037-4] [Citation(s) in RCA: 20] [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: 02/03/2021] [Accepted: 05/28/2021] [Indexed: 02/07/2023] Open
Abstract
Peripheral T-cell lymphoma (PTCL) is a heterogeneous group of non-Hodgkin lymphomas with poor prognosis. Up to 30% of PTCL lack distinctive features and are classified as PTCL, not otherwise specified (PTCL-NOS). To further improve our understanding of the genetic landscape and biology of PTCL-NOS, we perform RNA-sequencing of 18 cases and validate results in an independent cohort of 37 PTCL cases. We identify FYN-TRAF3IP2, KHDRBS1-LCK and SIN3A-FOXO1 as new in-frame fusion transcripts, with FYN-TRAF3IP2 as a recurrent fusion detected in 8 of 55 cases. Using ex vivo and in vivo experiments, we demonstrate that FYN-TRAF3IP2 and KHDRBS1-LCK activate signaling pathways downstream of the T cell receptor (TCR) complex and confer therapeutic vulnerability to clinically available drugs.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Cell Line, Tumor
- Cell Membrane/metabolism
- Cohort Studies
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Forkhead Box Protein O1/genetics
- Forkhead Box Protein O1/metabolism
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Intracellular Signaling Peptides and Proteins/metabolism
- Kaplan-Meier Estimate
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism
- Lymphoma, T-Cell, Peripheral/genetics
- Lymphoma, T-Cell, Peripheral/metabolism
- Lymphoma, T-Cell, Peripheral/pathology
- Mice
- Mice, Inbred C57BL
- NF-kappa B/metabolism
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Proto-Oncogene Proteins c-fyn/genetics
- Proto-Oncogene Proteins c-fyn/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- RNA-Seq
- Receptors, Antigen, T-Cell/metabolism
- Signal Transduction/genetics
- Sin3 Histone Deacetylase and Corepressor Complex/genetics
- Sin3 Histone Deacetylase and Corepressor Complex/metabolism
- bcl-X Protein/antagonists & inhibitors
- bcl-X Protein/metabolism
Collapse
Affiliation(s)
- Koen Debackere
- Laboratory for Experimental Hematology, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
| | - Lukas Marcelis
- Translational Cell & Tissue Research, KU Leuven, Leuven, Belgium
| | - Sofie Demeyer
- Center for Cancer Biology, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Marlies Vanden Bempt
- Laboratory for Experimental Hematology, KU Leuven, Leuven, Belgium
- Center for Cancer Biology, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Nicole Mentens
- Center for Cancer Biology, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Olga Gielen
- Center for Cancer Biology, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Kris Jacobs
- Center for Cancer Biology, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Michael Broux
- Center for Cancer Biology, VIB, Leuven, Belgium
- Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Gregor Verhoef
- Laboratory for Experimental Hematology, KU Leuven, Leuven, Belgium
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium
| | - Lucienne Michaux
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Carlos Graux
- Mont-Godinne University Hospital, Yvoir, Belgium
| | - Iwona Wlodarska
- Center for Human Genetics, KU Leuven, Leuven, Belgium
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Philippe Gaulard
- Département de Pathologie, Groupe Hospitalier Henri Mondor, AP-HP, Créteil, France
- INSERM U955 and Université Paris-Est, Créteil, France
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Thomas Tousseyn
- Translational Cell & Tissue Research, KU Leuven, Leuven, Belgium
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Jan Cools
- Center for Cancer Biology, VIB, Leuven, Belgium.
- Center for Human Genetics, KU Leuven, Leuven, Belgium.
| | - Daan Dierickx
- Laboratory for Experimental Hematology, KU Leuven, Leuven, Belgium.
- Department of Hematology, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|