1
|
Xie Z, Xia Y, Li Z, Zhang M, Tan Y, Han Y, Wang M, Zhang P, Li J. Integrated bioinformatics and functional studies identify CDK9 as a potential prognostic biomarker and therapeutic target in AML. Discov Oncol 2025; 16:1038. [PMID: 40490668 DOI: 10.1007/s12672-025-02841-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 05/28/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a highly heterogeneous disease characterized by complex genetic and molecular features that contribute to poor prognosis and low cure rates. Therefore, identifying novel therapeutic targets is crucial for improving treatment efficacy and patient survival. This study investigated the potential role of cyclin-dependent kinase 9 (CDK9), a known regulator of gene expression, in AML pathogenesis and prognosis. METHODS This study employed multiple bioinformatics approaches, including analysis of CDK9 expression across various cancers using the Tumor Immune Estimation Resource (TIMER2.0) database and further investigation of its expression and prognostic significance in AML using data from the Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. Survival analysis and Cox regression analysis were used to assess the association between CDK9 expression and patient prognosis. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were performed to elucidate the pathways and biological processes influenced by CDK9. Furthermore, the relationship between CDK9 expression and tumor immune infiltration was evaluated, and a protein-protein interaction (PPI) network was constructed. In vitro experiments, including Western blotting, CCK-8 assays, and flow cytometry, were conducted to validate the bioinformatics findings. RESULTS Bioinformatics analysis revealed significantly elevated CDK9 expression in AML samples, which correlated with poor patient prognosis. Functional enrichment analysis indicated that CDK9 is involved in key pathways related to cell proliferation, differentiation, and the tumor microenvironment. Moreover, the study observed a strong correlation between CDK9 expression and altered immune cell infiltration, suggesting a potential role in immune evasion. In vitro experiments confirmed that CDK9 overexpression promoted AML cell proliferation and inhibited apoptosis. Additionally, CDK9 showed a strong correlation with epithelial-mesenchymal transition (EMT)-related proteins, suggesting a potential role in AML progression and the EMT process. CONCLUSIONS This study demonstrates that CDK9 is a potential prognostic biomarker and therapeutic target in AML. Its involvement in multiple key pathways during AML development and its influence on the tumor immune microenvironment support further exploration of CDK9-targeted therapies to improve AML treatment outcomes.
Collapse
Affiliation(s)
- Zhibin Xie
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China
| | - Yang Xia
- Department of Finance, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Zhongyu Li
- Department of Hematology, The Third People's Hospital of Bengbu, Bengbu, Anhui, China
| | - Mengmeng Zhang
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China
| | - Yuanyuan Tan
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China
| | - Yuqing Han
- Bengbu Medical University, Bengbu, Anhui, China
| | - Meng Wang
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China
| | - Pingping Zhang
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China
| | - Jiajia Li
- Department of Hematology, First Affiliated Hospital of Bengbu Medical University, Bengbu, 233003, Anhui, China.
| |
Collapse
|
2
|
Zhang W, Kong D, Zhang X, Hu L, Nian Y, Shen Z. T cell aging and exhaustion: Mechanisms and clinical implications. Clin Immunol 2025; 275:110486. [PMID: 40120658 DOI: 10.1016/j.clim.2025.110486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
T cell senescence and exhaustion represent critical aspects of adaptive immune system dysfunction, with profound implications for health and the development of disease prevention and therapeutic strategies. These processes, though distinct, are interconnected at the molecular level, leading to impaired effector functions and reduced proliferative capacity of T cells. Such impairments increase susceptibility to diseases and diminish the efficacy of vaccines and treatments. Importantly, T cell senescence and exhaustion can dynamically influence each other, particularly in the context of chronic diseases. A deeper understanding of the molecular mechanisms underlying T cell senescence and exhaustion, as well as their interplay, is essential for elucidating the pathogenesis of related diseases and restoring dysfunctional immune responses. This knowledge will pave the way for the development of targeted therapeutic interventions and strategies to enhance immune competence. This review aims to summarize the characteristics, mechanisms, and disease associations of T cell senescence and exhaustion, while also delineating the distinctions and intersections between these two states to enhance our comprehension.
Collapse
Affiliation(s)
- Weiqi Zhang
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Dejun Kong
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Xiaohan Zhang
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China.
| | - Lu Hu
- Tianjin Medical University First Central Clinical College, Tianjin, China.
| | - Yeqi Nian
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Science, Tianjin, China; Department of Kidney Transplant, Tianjin First Central Hospital, Tianjin, China.
| | - Zhongyang Shen
- School of Medicine, Nankai University, Tianjin, China; Research Institute of Transplant Medicine, Nankai University, Tianjin, China; Tianjin Key Laboratory for Organ Transplantation, Tianjin, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Science, Tianjin, China.
| |
Collapse
|
3
|
Li D, Shi Y, Yu S, Zhang B, Huang Z, Ling F, Mao X, Deng Y, Cai M, Luo W. NK cellular derived nanovesicles in tumor immunity. Mol Immunol 2025; 182:54-61. [PMID: 40174421 DOI: 10.1016/j.molimm.2025.03.018] [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: 12/09/2024] [Revised: 02/20/2025] [Accepted: 03/30/2025] [Indexed: 04/04/2025]
Abstract
Natural Killer (NK) cells are a vital element of the innate immune system, and NK cell-based therapies have demonstrated efficacy against various malignancies. However, targeting solid tumors has been challenging due to the low infiltration of NK cells into tumors and the effective evasion strategies employed by tumors. Recent studies have shown that NK cell derived nanovesicles (NK-NV) can not only replicate the functions of NK cells but also offer more advantages in clinical applications. They are capable of transporting various cellular components such as proteins, nucleic acids, and lipids across distances, thereby facilitating intercellular communication among various cells within the tumor microenvironment (TME). With the progress in nanomedical technology, these vesicles can be engineered to carry a range of functional elements and therapeutic agents to enhance their antitumoral capabilities. In this review, we summarize the current available literature on NK-NVs, discuss their potential biological functions and the role of non-coding RNAs (ncRNAs), and explore their application in the treatment of solid tumors.
Collapse
Affiliation(s)
- Dingru Li
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Yixin Shi
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China; Liaoning Laboratory of Cancer Genomics and Epigenomics, Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Sifei Yu
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China
| | - Beiying Zhang
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China
| | - Ziyi Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Fei Ling
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiaofan Mao
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China
| | - Yuhua Deng
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China
| | - Mengyun Cai
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China.
| | - Wei Luo
- Institute of Translational Medicine, The First People's Hospital of Foshan, Guangdong, China.
| |
Collapse
|
4
|
Huo Y, Kim J, Kacanek D, Samer S, Livingston EG, Machado ES, Martinelli E. Antiretroviral Therapy at Conception Leads to Lower Peripheral CD49a + NK Cells and Higher SERPINB2. J Immunol Res 2025; 2025:4771787. [PMID: 40438469 PMCID: PMC12119168 DOI: 10.1155/jimr/4771787] [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: 01/24/2025] [Accepted: 04/30/2025] [Indexed: 06/01/2025] Open
Abstract
Problem: Antiretroviral therapy (ART) during pregnancy is essential to prevent vertical HIV transmission and preserve the health of the mother and child. However, ART in pregnancy has been associated with adverse birth outcomes linked to poor placental development. Immune dysregulation of placental development is an important factor in the development of preeclampsia (PE), a common hypertension disorder of pregnancy. Some studies found an association between ART use at conception or during the first trimester and PE. However, little is known regarding the impact of timing of ART initiation on the immune environment in pregnancy. Methods: To investigate the immune environment in pregnant persons with HIV (PPWH) on ART at conception (N = 40) compared to PPWH that started ART in the second trimester (N = 40) we analyzed specimens from the International Maternal Pediatric Adolescent AIDS Clinical Trials (IMPAACT) Perinatal Core Protocol, P1025, concluded in 2013. Results: No difference was found in soluble factors in circulation including PlGF and sFLt-1, associated with PE. However, upon analysis of PBMC by high dimension flow cytometry, we detected a lower frequency of circulating CD49a+ NK cells, associated with uterine tissue and pregnancy, in PPWH on ART at conception compared with PPWH who started ART in the second trimester. Moreover, PBMC from PPWH on ART at conception expressed higher levels of SERPINB2 in transcriptomics analyses. Conclusions: Our findings shed new insights into the potential impact of ART at conception and suggest the persistence of a dysregulated inflammatory environment compared to PPWH starting ART after the conclusion of placental development.
Collapse
Affiliation(s)
- Yanling Huo
- Center for Biostatistics in AIDS Research (CBAR), Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jinhee Kim
- Department of Medicine Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Deborah Kacanek
- Center for Biostatistics in AIDS Research (CBAR), Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Sadia Samer
- Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Elizabeth G. Livingston
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Elizabeth Stankiewicz Machado
- Department of Infectious Disease, University Hospital and Instituto De Pediatria Martagão Gesteira (IPPMG)-Federal University of Rio De Janeiro, Rio De Janeiro, Brazil
| | - Elena Martinelli
- Department of Medicine Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
5
|
Saeed AF. Tumor-Associated Macrophages: Polarization, Immunoregulation, and Immunotherapy. Cells 2025; 14:741. [PMID: 40422244 DOI: 10.3390/cells14100741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/28/2025] Open
Abstract
Tumor-associated macrophages' (TAMs) origin, polarization, and dynamic interaction in the tumor microenvironment (TME) influence cancer development. They are essential for homeostasis, monitoring, and immune protection. Cells from bone marrow or embryonic progenitors dynamically polarize into pro- or anti-tumor M2 or M1 phenotypes based on cytokines and metabolic signals. Recent advances in TAM heterogeneity, polarization, characterization, immunological responses, and therapy are described here. The manuscript details TAM functions and their role in resistance to PD-1/PD-L1 blockade. Similarly, TAM-targeted approaches, such as CSF-1R inhibition or PI3Kγ-driven reprogramming, are discussed to address anti-tumor immunity suppression. Furthermore, innovative biomarkers and combination therapy may enhance TAM-centric cancer therapies. It also stresses the relevance of this distinct immune cell in human health and disease, which could impact future research and therapies.
Collapse
|
6
|
Zhou Q, Siegelin MD, Greene LA. Targeting ATF5, CEBPB, and CEBPD with Cell-Penetrating Dpep Sensitizes Tumor Cells to NK-92MI Cell Cytotoxicity. Cells 2025; 14:667. [PMID: 40358191 PMCID: PMC12071554 DOI: 10.3390/cells14090667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/25/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Natural killer (NK) cells are an important innate defense against malignancies, and exogenous sources of NK cells have been developed as anti-cancer agents. Nevertheless, the apparent limitations of NK cells in clearing cancers have suggested that their efficacy might be augmented by combination with other treatments. We have developed cell-penetrating peptides that target the transcription factors ATF5, CEBPB, and CEBPD and that promote apoptotic cancer cell death both in vitro and in vivo without apparent toxicity to non-transformed cells. We report here that one such peptide, Dpep, significantly sensitizes a variety of tumor cell types to the cytotoxic activity of the NK cell line, NK-92MI. Such sensitization requires pre-exposure of tumor cells to Dpep and does not appear due to effects of Dpep on NK cells themselves. Our findings suggest that Dpep acts in this context to lower the apoptotic threshold of tumor cells to NK cell toxicity. Additionally, while Dpep pre-treatment does not prevent tumor cells from causing NK cell "inactivation", it sensitizes cancer cells to repeated rounds of exposure to fresh NK cells. These findings thus indicate that Dpep pre-treatment is an effective strategy to sensitize cancer cells to the cytotoxic actions of NK cells.
Collapse
Affiliation(s)
| | | | - Lloyd A. Greene
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA; (Q.Z.); (M.D.S.)
| |
Collapse
|
7
|
Lyu Z, Niu S, Fang Y, Chen Y, Li YR, Yang L. Addressing graft-versus-host disease in allogeneic cell-based immunotherapy for cancer. Exp Hematol Oncol 2025; 14:66. [PMID: 40317083 PMCID: PMC12046680 DOI: 10.1186/s40164-025-00654-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/13/2025] [Indexed: 05/04/2025] Open
Abstract
Allogeneic cell-based immunotherapies, particularly CAR-T cell therapy, represent a significant advancement in cancer treatment, offering scalable and consistent alternatives to autologous therapies. However, their widespread use is limited by the risk of graft-versus-host disease (GvHD). This review provides a comprehensive overview of GvHD in the context of allogeneic cell-based cancer immunotherapy and evaluates current strategies to mitigate its effects. Key strategies include genetic engineering approaches such as T cell receptor (TCR) knockout (KO) and T cell receptor alpha constant (TRAC) CAR knock-in. Alternative immune cell types like natural killer (NK) cells and natural killer T (NKT) cells offer potential solutions due to their lower alloreactivity. Additionally, stem cell technology, utilizing induced pluripotent stem cells (iPSCs), enables standardized and scalable production of engineered CAR-T cells. Clinical trials evaluating these strategies, such as UCART19 and CTX110, demonstrate promising results in preventing GvHD while maintaining anti-tumor efficacy. The review also addresses manufacturing considerations for allogeneic cell products and the challenges in translating preclinical findings into clinical success. By addressing these challenges, allogeneic cell-based immunotherapy continues to advance, paving the way for more accessible, scalable, and effective cancer treatments.
Collapse
Affiliation(s)
- Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Siyue Niu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Yuning Chen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA.
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, CA, 90095, USA.
- Goodman-Luskin Microbiome Center, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
8
|
Imani S, Farghadani R, Roozitalab G, Maghsoudloo M, Emadi M, Moradi A, Abedi B, Jabbarzadeh Kaboli P. Reprogramming the breast tumor immune microenvironment: cold-to-hot transition for enhanced immunotherapy. J Exp Clin Cancer Res 2025; 44:131. [PMID: 40281554 PMCID: PMC12032666 DOI: 10.1186/s13046-025-03394-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
This review discusses reprogramming the breast tumor immune microenvironment from an immunosuppressive cold state to an immunologically active hot state. A complex interplay is revealed, in which the accumulation of metabolic byproducts-such as lactate, reactive oxygen species (ROS), and ammonia-is shown to impair T-cell function and promote tumor immune escape. It is demonstrated that the tumor microenvironment (TME) is dominated by immunosuppressive cytokines, including interleukin-10 (IL-10), transforming growth factorβ (TGFβ), and IL-35. Notably, IL-35 is produced by regulatory T cells and breast cancer cells. The conversion of conventional T cells into IL-35-producing induced regulatory T cells, along with the inhibition of pro-inflammatory cytokine secretion, contributes to the suppression of anti-tumor immunity. It is further demonstrated that key immune checkpoint molecules-such as PD-1, PDL1, CTLA-4, TIM-3, LAG-3, and TIGIT-are upregulated within the TME, leading to Tcell exhaustion and diminished immune responses. The blockade of these checkpoints is shown to restore T-cell functionality and is proposed as a strategy to convert cold tumors into hot ones with robust effector cell infiltration. The therapeutic potential of chimeric antigen receptor (CAR)T cell therapy is also explored, and targeting specific tumor-associated antigens, such as glycoproteins and receptor tyrosine kinases, is highlighted. It is suggested that CART cell efficacy can be enhanced by combining these cells with immune checkpoint inhibitors and other immunomodulatory agents, thereby overcoming the barriers imposed by the immunosuppressive TME. Moreover, the role of the microbiome in regulating estrogen metabolism and systemic inflammation is reviewed. Alterations in the gut microbiota are shown to affect the TME, and microbiome-based interventions are proposed as an additional means to facilitate the cold-to-hot transition. It is concluded that by targeting the metabolic and immunological pathways that underpin immune suppression-through combination strategies involving checkpoint blockade, CART cell therapies, and microbiome modulation-the conversion of the breast TME from cold to hot can be achieved. This reprogramming is anticipated to enhance immune cell infiltration and function, thereby improving the overall efficacy of immunotherapies and leading to better clinical outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China.
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor Darul Ehsan, Malaysia
| | - Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Mahdieh Emadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Atefeh Moradi
- Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Behnaz Abedi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Parham Jabbarzadeh Kaboli
- Department of Biochemistry, Faculty of Medicine, Medical University of Warsaw, Warsaw, 02-091, Poland.
| |
Collapse
|
9
|
Xu Y, Wang T, Liang X, Yang J, Zhang Y, Bao S. Global research trends and focus on immunotherapy for endometrial cancer: a comprehensive bibliometric insight and visualization analysis (2012-2024). Front Immunol 2025; 16:1571800. [PMID: 40264788 PMCID: PMC12011754 DOI: 10.3389/fimmu.2025.1571800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Background This study conducted a novel systematic bibliometric and visualization analysis of global literature on immunotherapy for endometrial cancer (EC) to explore dynamic trends, research hotspots, and emerging topics, providing valuable references for future research. Methods Articles and reviews on EC immunotherapy published between 2012 and August 2024 were retrieved from the Web of Science Core Collection (WoSCC). Bibliometric tools, CiteSpace and VOSviewer, were used to analyze clustering patterns and research dynamics. Results A total of 861 articles were contributed by 5,331 authors from 1,392 institutions across 58 countries or regions, involving 1,823 keywords. China demonstrated outstanding performance in this field, contributing over 40% of the total publications and ranking first in publication volume. However, the total citation counts for publications from China lags that of the United States, highlighting the latter's leading position and areas for further improvement in China's research efforts. The University of Texas Medical Anderson Cancer Center and Nanjing Medical University were the two institutions with the highest number of publications. In terms of authorship, research teams led by Bosse, Tjalling, and Creutzberg, Carien L made significant contributions to advancing the field. Among individual publications, the work by Talhouk et al. achieved the highest average annual citation count of 70.88, demonstrating its profound impact. In terms of journals, Gynecologic Oncology emerged as a pivotal academic platform, publishing numerous articles and achieving the highest co-citation frequency. Additionally, Frontiers in Oncology, Frontiers in Immunology, and Frontiers in Genetics have become some of the most active and rapidly developing journals in recent years. Research hotspots are concentrated on themes such as the "Tumor Immune Microenvironment", "Immune Checkpoint Inhibitors", and "Targeted Therapy". Recent trends and frontier research focus on the combined application of immune checkpoint inhibitors with other therapies, research on the application of nanotechnology in immunotherapy, and the integration of artificial intelligence to enhance precision medicine. Additionally, efforts are increasingly directed toward advancing various immunotherapy strategies from basic research to clinical applications. Conclusions This comprehensive analysis reveals rapid advancements and significant potential in EC immunotherapy. Strengthening international collaboration and addressing barriers in the translation of research to clinical practice will drive further progress in this promising field.
Collapse
Affiliation(s)
- Yachen Xu
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Tao Wang
- School of Public Health, Hainan Medical University, Haikou, China
| | - Xiaojing Liang
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Jie Yang
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Yuxiang Zhang
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Shan Bao
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| |
Collapse
|
10
|
Nishida A, Andoh A. The Role of Inflammation in Cancer: Mechanisms of Tumor Initiation, Progression, and Metastasis. Cells 2025; 14:488. [PMID: 40214442 PMCID: PMC11987742 DOI: 10.3390/cells14070488] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Inflammation is an essential component of the immune response that protects the host against pathogens and facilitates tissue repair. Chronic inflammation is a critical factor in cancer development and progression. It affects every stage of tumor development, from initiation and promotion to invasion and metastasis. Tumors often create an inflammatory microenvironment that induces angiogenesis, immune suppression, and malignant growth. Immune cells within the tumor microenvironment interact actively with cancer cells, which drives progression through complex molecular mechanisms. Chronic inflammation is triggered by factors such as infections, obesity, and environmental toxins and is strongly linked to increased cancer risk. However, acute inflammatory responses can sometimes boost antitumor immunity; thus, inflammation presents both challenges and opportunities for therapeutic intervention. This review examines how inflammation contributes to tumor biology, emphasizing its dual role as a critical factor in tumorigenesis and as a potential therapeutic target.
Collapse
Affiliation(s)
- Atsushi Nishida
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Shiga, Japan;
| | | |
Collapse
|
11
|
Luo J, Guo M, Huang M, Liu Y, Qian Y, Liu Q, Cao X. Neoleukin-2/15-armored CAR-NK cells sustain superior therapeutic efficacy in solid tumors via c-Myc/NRF1 activation. Signal Transduct Target Ther 2025; 10:78. [PMID: 40025022 PMCID: PMC11873268 DOI: 10.1038/s41392-025-02158-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/22/2024] [Accepted: 01/22/2025] [Indexed: 03/04/2025] Open
Abstract
Adoptive transfer of chimeric antigen receptor (CAR)-modified natural killer (NK) cells represents a transformative approach that has significantly advanced clinical outcomes in patients with malignant hematological conditions. However, the efficacy of CAR-NK cells in treating solid tumors is limited by their exhaustion, impaired infiltration and poor persistence in the immunosuppressive tumor microenvironment (TME). As NK cell functional states are associated with IL-2 cascade, we engineered mesothelin-specific CAR-NK cells that secrete neoleukin-2/15 (Neo-2/15), an IL-2Rβγ agonist, to resist immunosuppressive polarization within TME. The adoptively transferred Neo-2/15-armored CAR-NK cells exhibited enhanced cytotoxicity, less exhaustion and longer persistence within TME, thereby having superior antitumor activity against pancreatic cancer and ovarian cancer. Mechanistically, Neo-2/15 provided sustained and enhanced downstream IL-2 receptor signaling, which promotes the expression of c-Myc and nuclear respiratory factor 1 (NRF1) in CAR-NK cells. This upregulation was crucial for maintaining mitochondrial adaptability and metabolic resilience, ultimately leading to increased cytotoxicity and pronounced persistence of CAR-NK cells within the TME. The resistance against TME immunosuppressive polarization necessitated the upregulation of NRF1, which is essential to the augmentative effects elicited by Neo-2/15. Overexpression of NRF1 significantly bolsters the antitumor efficacy of CAR-NK cells both in vitro and in vivo, with increased ATP production. Collectively, Neo-2/15-expressing CAR-NK cells exerts superior antitumor effects by exhaustion-resistance and longer survival in solid tumors.
Collapse
Affiliation(s)
- Jianhua Luo
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Meng Guo
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China.
| | - Mingyan Huang
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China
| | - Yanfang Liu
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Yuping Qian
- Department of Pathology, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Qiuyan Liu
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China.
| | - Xuetao Cao
- National Key Laboratory of Immunity & Inflammation, Institute of Immunology, Navy Medical University, Shanghai, 200433, China.
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China.
- Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
12
|
Xue Y, Wang Q, Chen Y, Zhang X, Tang J, Liu Y, Liu J. Biomimetic Diselenide-Sonosensitizer Nanoplatform for Enhanced Sonodynamic Therapy and In Situ Remodeling Immunosuppressive Microenvironment via Activating Innate and Adaptive Immunotherapy. Adv Healthc Mater 2025; 14:e2403998. [PMID: 39790035 DOI: 10.1002/adhm.202403998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Sonodynamic therapy (SDT), which is non-invasive and controllable has the potential to treat triple-negative breast cancer (TNBC). However, the hypoxia and immunosuppressive tumor microenvironment (TME) often block the production of reactive oxygen species and the induction of SDT-activated immunogenic cell death, thus limiting the activation of adaptive immune responses. To alleviate these challenges, we proposed the development of a multifunctional biomimetic nanoplatform (mTSeIR), which was designed with diselenide-conjugated sonosensitizers and tirapazamine (TPZ), encapsulated within M1 macrophage membrane. This nanoplatform utilized hypoxia-induced chemotherapy to improve the efficacy of SDT, to further enhance adaptive immunotherapy by activating innate immunity and remodeling the immunosuppressive TME. Firstly, the prodrug TPZ was activated due to the increased oxygen consumption associated with SDT. Subsequently, the mTSeIR enhanced repolarization of M2 macrophages to the M1 phenotype. The diselenide component in mTSeIR effectively activated the natural killer cell-mediated antitumor innate immune response. Ultimately, in vivo studies indicated that mTSeIR+US with good biosafety achieved over 98% tumor inhibition and enhanced adaptive immunotherapy. This research presents an efficient approach that addressed the limitations of SDT and achieves simultaneous activation of both innate and adaptive immunotherapy, resulting in significant antitumor and anti-metastatic efficacy in TNBC.
Collapse
Affiliation(s)
- Yifan Xue
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Qingliang Wang
- Department of Hepatobiliary-Pancreatic-Splenic Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510630, China
| | - You Chen
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Xiaoge Zhang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Junjie Tang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yadong Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Jie Liu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| |
Collapse
|
13
|
Zhou J, Yan P, Ma W, Li J. Cytokine modulation and immunoregulation of uterine NK cells in pregnancy disorders. Cytokine Growth Factor Rev 2025; 81:40-53. [PMID: 39603954 DOI: 10.1016/j.cytogfr.2024.11.007] [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: 11/11/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
Uterine natural killer (uNK) cells play a pivotal role in promoting placental development and supporting maternal-fetal immune tolerance, primarily through cytokine regulation and growth factor production. While the importance of uNK cells in pregnancy is well-established, the mechanisms of their interactions with trophoblasts and contributions to various pregnancy complications remain incompletely understood. This review highlights recent advancements in understanding uNK cell functions, with a focus on cytokine production, growth factor secretion, and receptor-ligand interactions, particularly involving killer immunoglobulin-like receptors (KIR) and human leukocyte antigen-C (HLA-C). We explore how uNK cell dysfunction contributes to pregnancy complications, including preeclampsia, recurrent pregnancy loss, and placenta accreta spectrum (PAS) disorders, emphasizing their roles in immune tolerance and placental health. By detailing the distinct cytokine signaling pathways and functional subtypes of uNK cells, this review provides insights into their regulatory mechanisms essential for pregnancy maintenance. Additionally, we discuss emerging therapeutic strategies targeting uNK-trophoblast interactions and propose future research directions, including the development of non-invasive biomarkers and personalized interventions. This comprehensive review addresses critical knowledge gaps, aiming to advance research in reproductive immunology and guide therapeutic innovations in maternal health.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China.
| | - Ping Yan
- Qingdao City Health Care Center for Cadres, Qingdao, Shandong 266071, China.
| | - Wenxue Ma
- Division of Regenerative Medicine, Department of Medicine, Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA 92093, USA.
| | - Jing Li
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China.
| |
Collapse
|
14
|
Nair AP, Ghosh S, Babu VS, Praveen M, Xin Y, Sahu GR, Vaidya TA, Debnath J, Raja K, Gadde SGK, M B T, Shetty N, Saxena A, Shetty R, Hose S, Deshpande V, Chakrabarty K, Handa JT, Qian JJ, Sethu S, Sinha D, Ghosh A. Attenuated adenosine mediated immune-dampening increases natural killer cell activity in early age-related macular degeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634301. [PMID: 39975064 PMCID: PMC11838234 DOI: 10.1101/2025.01.22.634301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Non-exudative age-related macular degeneration (AMD) involves retinal pigment epithelium (RPE) dysfunction and has been linked to altered intraocular immunity. Our investigation focuses on immune cell subsets and inflammation-associated factors in the eyes with early and intermediate AMD. We observed elevated levels of activated natural killer (NK) cells and interferon-γ, concurrent with reduced myeloid-derived suppressor cells (MDSCs) and adenosine in AMD eyes. Aqueous humor from AMD patients had diminished ability to dampen NK cell activation, an effect rescued by adenosine supplementation. The Cryba1 cKO mouse model recapitulated these immune alterations, and single-cell RNA-sequencing identified NK cell-related genes and NK cell-RPE interactions. Co-culture of activated NK cells with RPE cells induced barrier dysfunction and Gasdermin-E driven pyroptosis providing a functional link relevant to AMD. These findings suggest a double-hit model where elevated immune activation and loss of immune dampening mechanisms drive AMD progression. Resetting the intraocular immune balance may be a promising therapeutic strategy for managing early and intermediate AMD.
Collapse
Affiliation(s)
- Archana Padmanabhan Nair
- GROW Research Laboratory, Narayana Netralaya Foundation, Bengaluru, Karnataka, India
- Manipal Academy of Higher Education, Manipal, India
| | - Sayan Ghosh
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vishnu Suresh Babu
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Machiraju Praveen
- GROW Research Laboratory, Narayana Netralaya Foundation, Bengaluru, Karnataka, India
| | - Ying Xin
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ganesh Ram Sahu
- GROW Research Laboratory, Narayana Netralaya Foundation, Bengaluru, Karnataka, India
| | - Tanuja Arun Vaidya
- GROW Research Laboratory, Narayana Netralaya Foundation, Bengaluru, Karnataka, India
- Manipal Academy of Higher Education, Manipal, India
| | - Jayasree Debnath
- GROW Research Laboratory, Narayana Netralaya Foundation, Bengaluru, Karnataka, India
| | - Karthik Raja
- GROW Research Laboratory, Narayana Netralaya Foundation, Bengaluru, Karnataka, India
| | | | | | - Naren Shetty
- Narayana Nethralaya, Bengaluru, Karnataka, India
| | | | - Rohit Shetty
- Narayana Nethralaya, Bengaluru, Karnataka, India
| | - Stacey Hose
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vrushali Deshpande
- GROW Research Laboratory, Narayana Netralaya Foundation, Bengaluru, Karnataka, India
| | - Koushik Chakrabarty
- GROW Research Laboratory, Narayana Netralaya Foundation, Bengaluru, Karnataka, India
| | - James T. Handa
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - J. Jiang Qian
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Swaminathan Sethu
- GROW Research Laboratory, Narayana Netralaya Foundation, Bengaluru, Karnataka, India
| | - Debasish Sinha
- The Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Netralaya Foundation, Bengaluru, Karnataka, India
| |
Collapse
|
15
|
Kou Y, Tian Y, Ha Y, Wang S, Sun X, Lv S, Luo B, Yang Y, Qin L. Comprehensive Sepsis Risk Prediction in Leukemia Using a Random Forest Model and Restricted Cubic Spline Analysis. J Inflamm Res 2025; 18:1013-1032. [PMID: 39867945 PMCID: PMC11766288 DOI: 10.2147/jir.s505813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/12/2025] [Indexed: 01/28/2025] Open
Abstract
Background Sepsis is a severe complication in leukemia patients, contributing to high mortality rates. Identifying early predictors of sepsis is crucial for timely intervention. This study aimed to develop and validate a predictive model for sepsis risk in leukemia patients using machine learning techniques. Methods This retrospective study included 4310 leukemia patients admitted to the Affiliated Hospital of Guangdong Medical University from 2005 to 2024, using 70% for training and 30% for validation. Feature selection was performed using univariate logistic regression, LASSO, and the Boruta algorithm, followed by multivariate logistic regression analysis. Seven machine learning models were constructed and evaluated using receiver operating characteristic (ROC) curves and decision curve analysis (DCA). Shapley additive explanations (SHAP) were applied to interpret the results, and restricted cubic spline (RCS) regression explored the nonlinear relationships between variables and sepsis risk. Furthermore, we examined the interactions among predictors to better understand their potential interrelationships. Results The random forest (RF) model outperformed all others, achieving an AUC of 0.765 in the training cohort and 0.700 in the validation cohort. Key predictors of sepsis identified by SHAP analysis included C-reactive protein (CRP), procalcitonin (PCT), neutrophil count (Neut), lymphocyte count (Lymph), thrombin time (TT), red blood cell count (RBC), total bile acid (TBA), and systolic blood pressure (SBP). RCS analysis revealed significant non-linear associations between CPR, PCT, Neut, Lymph, TT, RBC and SBP with sepsis risk. Pairwise correlation analysis further revealed interactions among these variables. Conclusion The RF model exhibited robust predictive power for sepsis in leukemia patients, providing clinicians with a valuable tool for early risk assessment and the optimization of treatment strategies.
Collapse
Affiliation(s)
- Yanqi Kou
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, People’s Republic of China
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
| | - Yuan Tian
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
- Department of Pathology, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
| | - Yanping Ha
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
- Department of Pathology, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
| | - Shijie Wang
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, People’s Republic of China
| | - Xiaobai Sun
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, People’s Republic of China
| | - Shuxin Lv
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, People’s Republic of China
| | - Botao Luo
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
- Department of Pathology, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
| | - Yuping Yang
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang City, Guangdong Province, People’s Republic of China
| | - Ling Qin
- Department of Hematology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, People’s Republic of China
| |
Collapse
|