1
|
Chelladurai M, Sagi‐Assif O, Ben‐Menachem S, Meshel T, Pasmanik‐Chor M, Izraely S, Hoon DSB, Witz IP. A heterodimer of hemoglobin identifies theranostic targets on brain-metastasizing melanoma cells. Int J Cancer 2025; 157:773-787. [PMID: 40285526 PMCID: PMC12178103 DOI: 10.1002/ijc.35458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/17/2025] [Accepted: 04/08/2025] [Indexed: 04/29/2025]
Abstract
Cancer microenvironments encompass both cancer-promoting and cancer-restraining factors. If these factors cancel each other, cancer dormancy may ensue. In search of microenvironmental factors that keep dormant lung-metastasizing neuroblastoma cells and brain-metastasizing melanoma cells (BMMC) in check, we identified the beta subunit of hemoglobin and a heterodimer of alpha and beta chains of hemoglobin (α/β dimer) in the lung and brain microenvironments, respectively, as anti-metastatic factors. A previous study demonstrated that the α/β dimer triggers programmed cell death of BMMC and downregulates the expression of BRD4, GAB2, and IRS2 proteins, which perform essential functions in tumorigenesis and progression. The working hypothesis of the present study is that in addition to its tumoricidal function, the α/β dimer serves as a pathfinder for the identification of therapy targets for BMMC. We, therefore, employed small-molecule inhibitors of Bromodomain-containing protein 4 (BRD4), GRB2-associated-binding protein 2 (GAB2), and Insulin receptor substrate 2 (IRS2) as potential anti-BMMC agents. A combination of sub-lethal concentrations of BRD4 and IRS2 inhibitors synergistically arrested BMMC at the subG1 phase of the cell cycle and killed more than 70% of BMMCs. The BRD4/IRS2 inhibitor cocktail (designated hereafter as BRIRi) moderated the malignancy of BMMC lines from four different human melanomas. Preliminary results suggest that the BRIRi modulated "cold" BMMC to "hot" ones. Among the top enriched functions of differentially expressed genes identified by RNAseq of BRIRi-treated versus control BMMC, TNF and apoptotic signaling pathways were observed. We propose that co-targeting BRD4 and IRS2 offers a promising approach for treating BMMC.
Collapse
Affiliation(s)
- Maharrish Chelladurai
- The Shmunis School of Biomedicine and Cancer ResearchGeorge S. Wise Faculty of Life Science, Tel Aviv UniversityTel AvivIsrael
| | - Orit Sagi‐Assif
- The Shmunis School of Biomedicine and Cancer ResearchGeorge S. Wise Faculty of Life Science, Tel Aviv UniversityTel AvivIsrael
| | - Shlomit Ben‐Menachem
- The Shmunis School of Biomedicine and Cancer ResearchGeorge S. Wise Faculty of Life Science, Tel Aviv UniversityTel AvivIsrael
| | - Tsipi Meshel
- The Shmunis School of Biomedicine and Cancer ResearchGeorge S. Wise Faculty of Life Science, Tel Aviv UniversityTel AvivIsrael
| | - Metsada Pasmanik‐Chor
- Bioinformatics UnitThe George S. Wise Faculty of Life Science, Tel Aviv UniversityTel‐AvivIsrael
| | - Sivan Izraely
- The Shmunis School of Biomedicine and Cancer ResearchGeorge S. Wise Faculty of Life Science, Tel Aviv UniversityTel AvivIsrael
| | - Dave S. B. Hoon
- Department of Translational Molecular Medicine and Sequencing CenterSaint John's Cancer Institute at Providence Saint John's Health CenterSanta MonicaCaliforniaUSA
| | - Isaac P. Witz
- The Shmunis School of Biomedicine and Cancer ResearchGeorge S. Wise Faculty of Life Science, Tel Aviv UniversityTel AvivIsrael
| |
Collapse
|
2
|
Xu J, Jia Z, Zhao X, Wang L, Jin G, Li Z, Yin N, Li Y, Peng M. BCOR and ZC3H12A suppress a core stemness program in exhausted CD8+ T cells. J Exp Med 2025; 222:e20241133. [PMID: 40327039 PMCID: PMC12054362 DOI: 10.1084/jem.20241133] [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: 07/02/2024] [Revised: 11/25/2024] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
In chronic viral infections, sustained CD8+ T cell response relies on TCF1+ precursor-exhausted T cells (TPEX) exhibiting stem-like properties. TPEX self-renew and respond to PD-1 blockade, underscoring their paramount importance. However, strategies for effectively augmenting TPEX remain limited. Here, we demonstrate that ZC3H12A deficiency initiates a stemness program in TPEX but also increases cell death, whereas BCOR deficiency predominantly promotes TPEX proliferation. Consequently, co-targeting of both BCOR and ZC3H12A imparts exceptional stemness and functionality to TPEX, thereby enhancing viral control. Mechanistically, BCOR and ZC3H12A collaboratively suppress a core stemness program in TPEX characterized by heightened expression of ∼216 factors. While TCF1 plays a role, this core stemness program relies on novel factors, including PDZK1IP1, IFIT3, PIM2, LTB, and POU2F2. Crucially, overexpressing POU2F2 robustly boosts TPEX and enhances antiviral immunity. Thus, a core stemness program exists in exhausted T cells, jointly repressed by BCOR and ZC3H12A, robustly controlling TPEX differentiation and providing new targets for addressing T cell exhaustion.
Collapse
Affiliation(s)
- Jing Xu
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zeran Jia
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- IDG/McGovern Institute for Brain Research, State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Xiaocui Zhao
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Lixia Wang
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Gang Jin
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zhuoyang Li
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Na Yin
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Yinqing Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- IDG/McGovern Institute for Brain Research, State Key Laboratory of Molecular Oncology, MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Min Peng
- State Key Laboratory of Molecular Oncology, Institute for Immunology, Beijing Key Laboratory of Immunological Research of Allergy, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
3
|
Zhang Y, Fan Y, Hu Y, Wang X, Wen B, Duan X, Li H, Dong S, Yan Z, Zhang W, Jing Y. The role of MBD2 in immune cell development, function, and autoimmune diseases. Cell Death Discov 2025; 11:280. [PMID: 40533455 PMCID: PMC12177077 DOI: 10.1038/s41420-025-02563-0] [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: 03/08/2025] [Revised: 05/06/2025] [Accepted: 06/09/2025] [Indexed: 06/22/2025] Open
Abstract
DNA methylation is a key epigenetic modification that regulates gene expression, cell differentiation, and genome stability. Aberrant DNA methylation patterns, including the hypermethylation or global hypomethylation of tumor suppressor genes, are strongly associated with various human diseases, such as cancer, autoimmune disorders, and metabolic syndrome. DNA methylation predominantly occurs at CpG dinucleotides, influencing transcription by altering chromatin structure and accessibility. MBD2 (Methyl-CpG-binding proteins 2) play a crucial role in interpreting these epigenetic marks and regulating downstream gene expression. In disease contexts, aberrant DNA methylation disrupts cellular homeostasis by silencing key regulatory genes or activating pathological pathways. Current research primarily focuses on MBD2 in cancer, with less emphasis on its role in autoimmune diseases. This review discusses the role of MBD2 in regulating immune cell development and differentiation through epigenetic mechanisms, particularly DNA methylation and its regulatory components. Furthermore, it highlights the mechanistic contributions of MBD2 to autoimmune diseases such as systemic lupus erythematosus and evaluates its potential as a novel therapeutic target for these conditions.
Collapse
Affiliation(s)
- Yunfei Zhang
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yufeng Fan
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Ying Hu
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xiaocui Wang
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Bin Wen
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xuemei Duan
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Haonan Li
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Shumin Dong
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Ze Yan
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Weiwei Zhang
- Department of Anesthesiology, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China.
| | - Yukai Jing
- Department of Clinical Laboratory, Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China.
| |
Collapse
|
4
|
Wang H, Zhang N, Xu R, Ji C, Wei Y, Mi Q. The PD-1/PD-L1 pathway and Epstein-Barr virus. Eur J Med Res 2025; 30:486. [PMID: 40533842 PMCID: PMC12175326 DOI: 10.1186/s40001-025-02694-1] [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: 03/20/2025] [Accepted: 05/15/2025] [Indexed: 06/22/2025] Open
Abstract
Epstein-Barr virus (EBV) is a gamma-herpesvirus with double-stranded DNA. Primary EBV infection leads to infectious mononucleosis (IM) in 20-50% of children and young adults. EBV establishes latent infection in B lymphocytes and can infect T lymphocytes and NK cells, potentially causing lymphoproliferative disorders (LPD) and malignancies. While the PD-1/PD-L1 pathway's role in chronic viral infections is well-established, its specific functions in EBV infection remain poorly understood. Growing evidence suggests this pathway facilitates EBV immune evasion, yet the effect of PD-1 upregulation on Epstein-Barr virus-specific CD8 + T cell function during acute IM is unclear. Furthermore, the role of PD-1/PD-L1 pathway in cytotoxic T cells and immune regulation during EBV infection is still controversial. This review systematically analyzes current knowledge on PD-1/PD-L1 signaling in EBV infection, focusing on three key aspects: (1) its dual role in maintaining immune homeostasis during acute infection while potentially facilitating viral persistence, (2) its emerging potential as a diagnostic biomarker for disease progression and prognosis, particularly during acute infectious mononucleosis, and (3) the therapeutic implications of pathway modulation. We critically evaluate recent advances that position PD-1/PD-L1 at the intersection of virology and tumor immunology, while highlighting important unanswered questions that require further investigation to optimize EBV-specific immunotherapies.
Collapse
Affiliation(s)
- Hui Wang
- Department of Pediatrics, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Ning Zhang
- Department of Pediatrics, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Rongshuang Xu
- Department of Pediatrics, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Cundong Ji
- Department of Emergency, Taian 88 Hospital, Taian, Shandong, China
| | - Youzhen Wei
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Qing Mi
- Department of Pediatrics, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China.
| |
Collapse
|
5
|
Nennig K, Shaw T, Borsinger L, Bailey AL. Ineffectual immunity in a resurrected mouse model of persistent viremia. J Virol 2025; 99:e0024825. [PMID: 40338081 DOI: 10.1128/jvi.00248-25] [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: 02/09/2025] [Accepted: 04/07/2025] [Indexed: 05/09/2025] Open
Abstract
Viruses that establish persistent (i.e., chronic) infections have evolved sophisticated strategies to avoid clearance by the host immune system. This is particularly true for viruses that infect immunocompetent mammals and sustain high infectious burdens in body sites under intense immune surveillance (i.e., the blood, a.k.a., "viremia"). Historically, lymphocytic choriomeningitis virus (LCMV) infection of laboratory mice has served as a powerful model to understand mechanisms of failed immunity, but other viruses may have unique and underappreciated persistence strategies. Here, we resurrect a bygone model of viral persistence-lactate dehydrogenase-elevating virus (LDV)-and use modern transgenic mouse technologies to investigate various aspects of anti-viral immunity. We find that interferons have a modest impact on LDV replication, with interferon-alpha blunting LDV viremia in the acute phase of the infection and interferon-gamma reducing LDV viral loads in the chronic phase of infection, but only when paired with an intact interferon-alpha response. Adaptive immunity, assessed in Rag-knockout mice, had only a modest impact on LDV viremia, and only during the sub-acute phase of infection. Mice lacking the critical immune checkpoint molecule PD-1 showed no signs of disease and supported LDV viral loads at levels equivalent to their wild-type counterparts. Altogether, these results point to a novel and highly effective mechanism of persistence that is minimally impacted by conventional aspects of anti-viral immunity or immune exhaustion-a rarity among persistent viruses. Given the relative paucity of chronic infection models in the laboratory mouse, LDV infection may be useful for exploring unique modes of immune system failure. IMPORTANCE Viruses that infect a host over long periods of time have evolved unique strategies to evade the host immune system. Of particular interest are viruses that cause persistent infection in the laboratory mouse-the most well-developed tool for studying the mammalian immune system. Here, we resurrected a model of persistent RNA virus infection (lactate dehydrogenase-elevating virus, LDV) and applied modern tools of mouse immunology to further characterize its persistence. We found that host factors that typically have a dramatic effect on viral infections-e.g., the interferon system and lymphocytes-had very little impact on LDV infection. Removing "checks" on immune activation also had little effect on the virus or host health. Altogether, these findings imply that LDV uses a unique and highly effective mechanism to avoid immune clearance. Understanding this mechanism has implications for understanding ways in which the immune system fails.
Collapse
Affiliation(s)
- Kylie Nennig
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Teressa Shaw
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Logan Borsinger
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Adam L Bailey
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
6
|
Bargiel K, Scriabine I, Herr F, de Goër de Herve MG, Hendel-Chavez H, Taoufik Y, Fourati S, Dekeyser M, Durrbach A. BKPyV-specific T-cell Exhaustion in Kidney Transplant Recipients: Targeting Inhibitory Receptors Improves Their Functionality. Transplantation 2025:00007890-990000000-01104. [PMID: 40525862 DOI: 10.1097/tp.0000000000005448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2025]
Abstract
BACKGROUND BK polyomavirus (BKPyV) nephropathy is a significant complication of kidney transplantation associated with high levels of BKPyV replication in plasma and poor graft survival. It is currently treated by reducing immunosuppression to restore the immune response. METHODS We analyzed circulating T cells from 28 kidney transplant recipients with detectable levels of BKPyV DNA in the blood (BKPyV DNAemia). Immunosuppression was significantly reduced in all these patients. We evaluated BKPyV-specific T-cell functionality and phenotype and assessed graft outcomes prospectively. RESULTS BKPyV DNAemia was rapidly controlled in 13 patients (controllers [C] group), whereas viral replication was sustained in the other 15, who were considered not to have responded to reduced immunosuppression (noncontroller [NC] group). The induction and maintenance therapies used were similar in the C and NC groups. The slope of renal function decline tended to be worse in the NC group than in the C group (P < 0.055). BKPyV-specific T-cell functions (T-cell proliferation and cytokine secretion) were weaker in the NC group than in the C group. This functional impairment was associated with an overexpression of several inhibitory receptors (programmed cell death protein 1 [PD1], T-cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory motif domains, or T-cell immunoglobulin and mucin-containing protein 3 [TIM3]), highlighting an exhausted-like phenotype of BKPyV-specific CD4 and CD8 T cells in the NC group. T-cell inhibition was not overcome by a single blocking antibody against inhibitory receptors, whereas a combination of anti-PD1 and anti-TIM3 antibodies significantly restored BKPyV-specific CD8 T-cell functions (P < 0.05). CONCLUSIONS Sustained BKPyV DNAemia was associated with an exhausted phenotype of BKPyV-specific T cells despite immunosuppression reduction in kidney transplant recipients. We show that anti-BKPyV-specific CD8 functions can be restored ex vivo by blocking the PD1 and TIM3 pathways, paving the way for new treatment strategies.
Collapse
Affiliation(s)
- Karen Bargiel
- INSERM UMR1186, Immunologie intégrative des tumeurs et immunothérapie des cancers, Gustave Roussy Institute, Villejuif, France
- Faculté de Medecin Le Kremlin Bicetre, Paris-Saclay University, Paris, France
| | - Ivan Scriabine
- INSERM UMR1186, Immunologie intégrative des tumeurs et immunothérapie des cancers, Gustave Roussy Institute, Villejuif, France
- Faculté de Medecin Le Kremlin Bicetre, Paris-Saclay University, Paris, France
| | - Florence Herr
- INSERM UMR1186, Immunologie intégrative des tumeurs et immunothérapie des cancers, Gustave Roussy Institute, Villejuif, France
- Faculté de Medecin Le Kremlin Bicetre, Paris-Saclay University, Paris, France
| | - Marie-Ghislaine de Goër de Herve
- INSERM UMR1186, Immunologie intégrative des tumeurs et immunothérapie des cancers, Gustave Roussy Institute, Villejuif, France
- Faculté de Medecin Le Kremlin Bicetre, Paris-Saclay University, Paris, France
| | - Houria Hendel-Chavez
- INSERM UMR1186, Immunologie intégrative des tumeurs et immunothérapie des cancers, Gustave Roussy Institute, Villejuif, France
- Faculté de Medecin Le Kremlin Bicetre, Paris-Saclay University, Paris, France
| | - Yassine Taoufik
- INSERM UMR1186, Immunologie intégrative des tumeurs et immunothérapie des cancers, Gustave Roussy Institute, Villejuif, France
- Faculté de Medecin Le Kremlin Bicetre, Paris-Saclay University, Paris, France
| | - Slim Fourati
- Department of Virology, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Creteil, France
| | - Manon Dekeyser
- INSERM UMR1186, Immunologie intégrative des tumeurs et immunothérapie des cancers, Gustave Roussy Institute, Villejuif, France
- Faculté de Medecin Le Kremlin Bicetre, Paris-Saclay University, Paris, France
- Department of Nephrology, Centre Hospitalier Régional Universitaire d'Orléans, Orléans, France
| | - Antoine Durrbach
- INSERM UMR1186, Immunologie intégrative des tumeurs et immunothérapie des cancers, Gustave Roussy Institute, Villejuif, France
- Faculté de Medecin Le Kremlin Bicetre, Paris-Saclay University, Paris, France
- Department of Nephrology and Transplantation, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Creteil, France
| |
Collapse
|
7
|
Ji H, Zhang L, Ye L. Exosome, an important transmitter in the drug resistance of non-small cell lung cancer. Front Oncol 2025; 15:1539047. [PMID: 40444086 PMCID: PMC12119617 DOI: 10.3389/fonc.2025.1539047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
Recent studies have promoted new insights into the biology of non-small cell lung cancer (NSCLC) and made considerable progress in the field of treatment, including targeted therapy for driver gene mutations. Immunotherapy (IO) is another breakthrough, which has achieved amazing clinical efficacy. However, the survival status of advanced NSCLC patients is still unsatisfactory. Drug resistance is an urgent problem to be solved in almost all anti-cancer treatment schemes. Nowadays, platinum based chemotherapy remains the standard treatment for patients with driver gene negative advanced NSCLC. Previous studies have shown that the reduction of intracellular accumulation of platinum drugs, DNA damage repair and the enhancement of detoxification effect all lead to platinum resistance. The mechanisms of tyrosine kinase inhibitors (TKIs) resistance include the emergence of secondary mutation, the activation of bypass signal pathways, the abnormality of downstream signal pathways and the transformation of phenotype. The mechanisms of immune checkpoint inhibitors (ICIs) resistance are more complex. A variety of cells, cytokines and metabolites participate in it to form an immunosuppressive microenvironment, resulting in the impairment of effector T cell function. Exosomes are small molecules secreted by a variety of cells. They can carry information such as miRNA, lncRNA, and protein, and play a pivotal role in signal transduction between cells. More and more studies show that exosomes are important transmitters in lung cancer cells, which can transfer drug resistance information from drug-resistant cells to sensitive cells. However, the underling specific mechanisms need to be further explored to find a new breakthrough for overcoming drug resistance of NSCLC.
Collapse
Affiliation(s)
- Hongzhi Ji
- Department of Respiratory, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Li Zhang
- Department of Gastroenterology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, China
| | - Lingyun Ye
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Zheng K, Dai L, Zhang S, Zhao Y, Li W, Gao Y, Mang Y, Jiao L, Tang Y, Ran J. Unraveling the Heterogeneity of CD8+ T-Cell Subsets in Liver Cirrhosis: Implications for Disease Progression. Gut Liver 2025; 19:410-426. [PMID: 38623058 PMCID: PMC12070210 DOI: 10.5009/gnl230345] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/26/2023] [Accepted: 01/03/2024] [Indexed: 04/17/2024] Open
Abstract
Background/Aims : Liver cirrhosis involves chronic inflammation and progressive fibrosis. Among various immune cells, CD8+ T cells are considered a major contributor to hepatic inflammation and fibrosis. However, the exact molecular pathways governing CD8+ T-cell-mediated effects in cirrhosis remain unclear. Methods : This study analyzed transcriptomic and single-cell sequencing data to elucidate CD8+ T-cell heterogeneity and implications in cirrhosis. Results : Weighted gene co-expression analysis of bulk RNA-seq data revealed an association between cirrhosis severity and activated T-cell markers like HLA and chemokine genes. Furthermore, single-cell profiling uncovered eight CD8+ T-cell subtypes, notably, effector memory (Tem) and exhausted (Tex) T cells. Tex cells, defined by PDCD1, LAG3, and CXCL13 expression, were increased in cirrhosis, while Tem cells were decreased. Lineage tracing and differential analysis highlighted CXCL13+ Tex cells as a terminal, exhausted subtype of cells with roles in PD-1 signaling, glycolysis, and T-cell regulation. CXCL13+ Tex cells displayed T-cell exhaustion markers like PDCD1, HAVCR2, TIGIT, and TNFRSF9. Functional analysis implicated potential roles of these cells in immunosuppression. Finally, a CXCL13+ Tex-cell gene signature was found that correlated with cirrhosis severity and poorer prognosis of liver cancer. Conclusions : In summary, this comprehensive study defines specialized CD8+ T-cell subpopulations in cirrhosis, with CXCL13+ Tex cells displaying an exhausted phenotype associated with immune dysregulation and advanced disease. Key genes and pathways regulating these cells present potential therapeutic targets.
Collapse
Affiliation(s)
- Kepu Zheng
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Leiyang Dai
- Inspection Department of Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Shengning Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Yingpeng Zhao
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Wang Li
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Yang Gao
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Yuanyi Mang
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Lingfeng Jiao
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| | - Yu Tang
- Kunming Medical University, Kunming, China
| | - Jianghua Ran
- Department of Hepato-Biliary-Pancreatic Surgery, The First People's Hospital of Kunming, The Affiliated Calmette Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
9
|
Walter V, Koch A, Hillmann D, Dannehl D, Staebler A, Pfister K, Risch L, Engler T, Brucker S, Janni W, Hartkopf A, Flatz L. Association of CMV status with response to neoadjuvant chemoimmunotherapy in early triple-negative breast cancer. Cancer Lett 2025; 626:217789. [PMID: 40354994 DOI: 10.1016/j.canlet.2025.217789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 04/15/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Affiliation(s)
- Vincent Walter
- Department of Dermatology, University Hospital Tübingen, Germany
| | - André Koch
- Department of Women's Health, University Hospital Tübingen, Germany
| | | | - Dominik Dannehl
- Department of Women's Health, University Hospital Tübingen, Germany
| | | | | | - Lorenz Risch
- Division of Clinical Biochemistry, Medical University Innsbruck, Austrlia
| | - Tobias Engler
- Department of Women's Health, University Hospital Tübingen, Germany
| | - Sara Brucker
- Department of Women's Health, University Hospital Tübingen, Germany
| | - Wolfgang Janni
- Department of Gynecology, University Hospital Ulm, Germany
| | - Andreas Hartkopf
- Department of Women's Health, University Hospital Tübingen, Germany
| | - Lukas Flatz
- Department of Dermatology, University Hospital Tübingen, Germany; Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland.
| |
Collapse
|
10
|
Li Q, Xu S, Ren Y, Zhang C, Li K, Liu Y. Single-cell RNA sequencing reveals adrb1 as a sympathetic nerve-regulated immune checkpoint driving T cell exhaustion and impacting immunotherapy in esophageal squamous cell carcinoma. Front Immunol 2025; 16:1520766. [PMID: 40406147 PMCID: PMC12095256 DOI: 10.3389/fimmu.2025.1520766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 04/15/2025] [Indexed: 05/26/2025] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) presents significant health challenges due to its aggressive nature and poor prognosis from late-stage diagnosis. Despite these challenges, emerging therapies like immune checkpoint inhibitors offer hope. β1-adrenergic signaling has been implicated in T cell exhaustion, which weakens the immune response in ESCC. Blocking this pathway could restore T cell function. Recent advances in single-cell RNA sequencing (scRNA-seq) have enabled deeper insights into tumor heterogeneity and the immune landscape, opening the door for personalized treatment strategies that may improve survival and reduce resistance to therapy. Methods We combined scRNA-seq with bulk RNA analysis to explore adrenergic receptor signaling in ESCC, focusing on changes before and after neoadjuvant therapy. We identified ADRB1+ T cells through data analysis and experimental validation. Copy number variation (CNV) analysis detected malignant cells within scRNA-seq data, while intercellular interaction analysis examined communication between cell populations. Deconvolution of TCGA data revealed key immune populations, which were integrated into a prognostic model based on the adrenergic receptor signaling pathway and differentially expressed genes. Results The adrenergic receptor signaling pathway was found in various immune cells, including T cells. scRNA-seq analysis revealed increased ADRB1 expression in T cells after neoadjuvant therapy. Immunofluorescence confirmed colocalization of ADRB1 with T cells, and fluorescence-activated cell sorting (FACS) showed that ADRB1 expression was elevated alongside exhaustion markers, while immune function markers were reduced. CNV analysis highlighted malignant cells in the tumor microenvironment, and intercellular interaction analysis explored ADRB1+ T cells' role in immune support. Deconvolution of TCGA data identified ADRB1+ T cells, SPP1+ macrophages, and CD44+ malignant cells, all of which were prognostically significant. A prognostic model constructed from the intersection of the adrenergic receptor signaling pathway and differentially expressed genes following neoadjuvant therapy showed a significant prognostic effect. Conclusions ADRB1 expression increases after neoadjuvant therapy in ESCC and correlates with poor prognosis. Our findings suggest ADRB1 as a potential prognostic biomarker and therapeutic target for post-neoadjuvant immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Ying Liu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
11
|
Kakh M, Doroudchi M, Talepoor A. Induction of Regulatory T Cells After Virus Infection and Vaccination. Immunology 2025. [PMID: 40329764 DOI: 10.1111/imm.13927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 05/08/2025] Open
Abstract
Vaccines have been proven to be one of the safest and most effective ways to prevent and combat diseases. However, the main focus has been on the evaluation of the potency of effector mechanisms and the lack of adverse effects of vaccine candidates. Recently, the importance of induced regulatory mechanisms of the immune system after vaccination has come to light. With the increase in our knowledge about these regulatory mechanisms including the regulatory T cells (Tregs), we have come to understand the significance of this arm of the immune system in controlling immunopathology and/or diminishing the effectiveness of vaccines, especially viral vaccines. Tregs play a dual role during infectious diseases by limiting immune-mediated pathology and also contributing to chronic pathogen persistence by decreasing effector immunity and clearance of infection. Tregs may also affect immune responses after vaccination primarily by inhibiting antigen presenting cell function such as cytokine secretion and co-stimulatory molecule expression as well as effector T (Teff) and B cell function. In this article, we review the current knowledge on the induction of Tregs after several life-threatening virus infections and their available vaccines to bring them to the spotlight and emphasise that studying viral-induced antigen-specific Tregs will help us improve the effectiveness and decrease the immunopathology or side effects of viral vaccines. Trial Registration: ClinicalTrials.gov identifier: NCT04357444.
Collapse
Affiliation(s)
- MansourehKarimi Kakh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - AtefeGhamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Qureshi Z, Zaheer Z, Asghar Z, Bakhtiar M, Fatima E, Altaf F. Cardiovascular Risk Profile of Nivolumab Anti-cancer Therapy: A Review of Current Literature. Am J Clin Oncol 2025; 48:235-241. [PMID: 40008416 DOI: 10.1097/coc.0000000000001166] [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] [Indexed: 02/27/2025]
Abstract
OBJECTIVES Immune checkpoint inhibitors (ICI) upregulate host antitumor immunity, proving efficacy across diverse tumor types. Currently approved ICI treatment primarily targets the programmed cell death receptor 1 (PD-1) and its ligand PD-L1, and cytotoxic T lymphocyte-antigen 4 (CTLA-4). Nivolumab is a monoclonal antibody that targets the human PD-1 receptor and is an entirely human immunoglobulin G4 (IgG4), approved by the FDA for various cancers like advanced melanoma, metastatic renal cell carcinoma, Hodgkin lymphoma, and advanced lung carcinoma. This review will summarise and discuss the recent literature on cardiotoxicity associated with nivolumab therapy. METHODS We searched online databases like PubMed, Scopus, Google Scholar, and Embase for articles related to Nivolumab. RESULTS Cardiotoxicity with ICI use is most commonly represented as myocarditis. Patients present with complaints of shortness of breath, palpitations, edema, and fatigue. Takotsubo cardiomyopathy, or broken heart syndrome, is characterized by systolic dysfunction of the left ventricle, mimicking a myocardial infarction but without associated coronary ischemia and with minimal elevation of cardiac enzymes. In the CHECKMATE-037 trial, ventricular arrhythmias occurred in <10% of those who received nivolumab. In a retrospective analysis of patients treated with ICI (predominantly nivolumab monotherapy) for lung cancer, 11% of the patients developed major adverse cardiac events, including myocarditis, non-ST-segment elevated myocardial infarction, supraventricular tachycardia, and pericardial disorders. CONCLUSION Close collaboration between cardiology and oncology specialists is crucial for early detection and effective management of cardiac complications, enhancing the safety of nivolumab anticancer therapy.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | | | - Zoha Asghar
- Department of Medicine, Ziauddin University, Karachi
| | | | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| |
Collapse
|
13
|
Kolland D, Kuhlmann M, de Almeida GP, Köhler A, Arifovic A, von Strempel A, Pourjam M, Bolsega S, Wurmser C, Steiger K, Basic M, Neuhaus K, Schmidt-Weber CB, Stecher B, Zehn D, Ohnmacht C. A specific microbial consortium enhances Th1 immunity, improves LCMV viral clearance but aggravates LCMV disease pathology in mice. Nat Commun 2025; 16:3902. [PMID: 40274773 PMCID: PMC12022176 DOI: 10.1038/s41467-025-59073-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Anti-viral immunity can vary tremendously from individual to individual but mechanistic understanding is still scarce. Here, we show that a defined, low complex bacterial community (OMM12) but not the general absence of microbes in germ-free mice leads to a more potent immune response compared to the microbiome of specific-pathogen-free (SPF) mice after a systemic viral infection with LCMV Clone-13. Consequently, gnotobiotic mice colonized with OMM12 have more severe LCMV-induced disease pathology but also enhance viral clearance in the intestinal tract. Mechanistically, single-cell RNA sequencing analysis of adoptively transferred virus-specific T helper cells and endogenous T helper cells in the intestinal tract reveal a stronger pro-inflammatory Th1 profile and a more vigorous expansion in OMM12 than SPF mice. Altogether, our work highlights the causative function of the intestinal microbiome for shaping adaptive anti-viral immunity with implications for vaccination strategies and anti-cancer treatment regimens.
Collapse
Affiliation(s)
- Daphne Kolland
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Miriam Kuhlmann
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Gustavo P de Almeida
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Amelie Köhler
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Anela Arifovic
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Alexandra von Strempel
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
| | - Mohsen Pourjam
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Silvia Bolsega
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Christine Wurmser
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Klaus Neuhaus
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
- Member of the German Center of Lung Research (DZL), Partner Site Munich, Munich, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
- German Center for Infection Research (DZIF), partner site LMU, Munich, Germany
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany.
| |
Collapse
|
14
|
Meng D, Wang J, Du L, Hu X, Liu Y, Zhang P, Wang J, Dong Q. PD-L1 in plasmacytoid dendritic cells promote HBV persistence through disrupting humoral immune response. Front Immunol 2025; 16:1545667. [PMID: 40342414 PMCID: PMC12058763 DOI: 10.3389/fimmu.2025.1545667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 04/07/2025] [Indexed: 05/11/2025] Open
Abstract
Objective To investigate the efficacy of PD-L1 blockade in restoring humoral immune response against HBV. Methods HBV-persistent C57BL/6J mice were established through hydrodynamic tail vein injection of 10 µg pAAV-HBV1.2 plasmid. Subsequently, mice treated i.p. with anti-PD-L1 and/or anti-CTLA-4 at specified time points, with dosages of 500 µg, 250 µg, and 250 µg, respectively. Additionally, 5 × 105 magnetic bead-purified plasmacytoid dendritic cells (pDCs) were adoptively transferred i.v. into the acute mouse model followed by anti-PD-L1 treatment. Quantitative real-time PCR was employed to assess the expression levels of costimulatory and tolerogenic molecules in two dendritic cell subsets. Serum HBsAg and HBsAb were measured using ELISA. Flow cytometry was utilized to quantify T follicular helper (Tfh) cells, regulatory T cells (Treg), and germinal center (GC) B cells. Results PD-L1 blockade markedly enhanced the differentiation of Tfh cells and GC B cells in HBV-persistent C57BL/6J mice, thereby promoting HBV clearance. Additionally, pDCs exhibited an increased capacity to induce immune tolerance, with pDCs isolated from HBV carriers inducing viral persistence. This persistence was effectively counteracted by treatment with anti-PD-L1. Conclusion pDCs mediate the dysregulation of the humoral immune response to HBV through PD-L1 in chronic hepatitis B infection, highlighting a promising target for the management of chronic HBV.
Collapse
Affiliation(s)
- Danyang Meng
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, China
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, China
| | - Jinhao Wang
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, China
| | - Lianqun Du
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, China
| | - Xiaojun Hu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, China
| | - Ying Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, China
| | - Pengcheng Zhang
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, China
| | - Jianjie Wang
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Qingyang Dong
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin, China
| |
Collapse
|
15
|
Köksal H, Herbst M, Perreira P, Nater M, Regli N, Boudjeniba C, Erdem Borgoni N, Cecconi V, van den Broek M. Pre-existing intratumoral stem-like CD8 + T cells drive radiotherapy-induced tumor immunity. Cell Rep 2025; 44:115566. [PMID: 40215165 DOI: 10.1016/j.celrep.2025.115566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/19/2025] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
CD8+ T cells are crucial for both spontaneous and therapy-induced restriction of tumor progression. Although many patients with cancer undergo radiotherapy, the precise effect of this genotoxic treatment on tumor-associated CD8+ T cells is insufficiently understood. Here, we investigated the influence of radiotherapy on intratumoral CD8+ T cells. We found that, although these CD8+ T cells initially decline following radiotherapy, they subsequently expand and are both essential and sufficient for early tumor control. In response to radiotherapy, stem-like CD8+ T cells proliferate and differentiate into effector CD8+ T cells, making them key drivers of tumor immunity. Our findings underscore the pivotal role of intratumoral stem-like CD8+ T cells in mediating radiotherapy-induced anti-tumor immunity and provide deeper insights into the dynamic behavior of CD8+ T cells during tumor control after radiotherapy.
Collapse
Affiliation(s)
- Hakan Köksal
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Michael Herbst
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Paulo Perreira
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Marc Nater
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Nicola Regli
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Nese Erdem Borgoni
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Maries van den Broek
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
16
|
Saleem M, Watson AE, Anwaar A, Jasser AO, Yusuf N. Optimizing Immunotherapy: The Synergy of Immune Checkpoint Inhibitors with Artificial Intelligence in Melanoma Treatment. Biomolecules 2025; 15:589. [PMID: 40305346 PMCID: PMC12024560 DOI: 10.3390/biom15040589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/01/2025] [Accepted: 04/12/2025] [Indexed: 05/02/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed melanoma treatment; however, predicting patient responses remains a significant challenge. This study reviews the potential of artificial intelligence (AI) to optimize ICI therapy in melanoma by integrating various diagnostic tools. Through a comprehensive literature review, we analyzed studies on AI applications in melanoma immunotherapy, focusing on predictive modeling, biomarker identification, and treatment response prediction. Key findings highlight the efficacy of AI in improving ICI outcomes. Machine learning models successfully identified prognostic cytokine signatures linked to nivolumab clearance. The combination of AI with RNAseq analysis had the potential for the development of personalized treatment with ICIs. A machine learning-based approach was able to assess the risk-benefit ratio for the prediction of immune-related adverse events (irAEs) using the electronic health record (EHR) data. Deep learning algorithms demonstrated high accuracy in tumor microenvironment analysis, including tumor region identification and lymphocyte detection. AI-assisted quantification of tumor-infiltrating lymphocytes (TILs) proved prognostically valuable in primary melanoma and predictive of anti-PD-1 therapy response in metastatic cases. Integrating multiple diagnostic modalities, such as CT imaging and laboratory data, modestly enhanced predictive performance for 1-year survival in advanced cancers treated with immunotherapy. These findings underscore the potential of AI-driven approaches to refine biomarker identification, treatment prediction, and patient stratification in melanoma immunotherapy. While promising, clinical validation and implementation challenges remain.
Collapse
Affiliation(s)
- Mohammad Saleem
- Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA (A.A.); (A.O.J.)
| | - Abigail E. Watson
- College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Aisha Anwaar
- Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA (A.A.); (A.O.J.)
| | - Ahmad Omar Jasser
- Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA (A.A.); (A.O.J.)
| | - Nabiha Yusuf
- Department of Dermatology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA (A.A.); (A.O.J.)
| |
Collapse
|
17
|
Córdoba-Lanús E, García-Pérez O, Melgar-Vilaplana L, Domínguez-de-Barros A, Fernández-de-Misa R. Germline PDCDL1 Gene Variants Are Associated with Increased Primary Melanoma Thickness. Biomolecules 2025; 15:584. [PMID: 40305358 PMCID: PMC12024702 DOI: 10.3390/biom15040584] [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: 03/06/2025] [Revised: 04/05/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025] Open
Abstract
Background: The incidence of malignant melanoma (MM) continues to increase annually, and tumour invasiveness is a main prognostic factor. Single-nucleotide polymorphisms (SNPs) have become key tools in the study of cancer genetics, influencing susceptibility and prognosis. Methods: In the present study, we analysed the relationship between five SNPs on the PDCDL1 gene (rs822336, rs822337, rs822338, rs229736, rs4143815) with prognosis as well as primary tumour invasiveness characteristics in 377 whole blood samples from MM individuals. Results: Patients who presented the rs822336 CG or GG genotypes (OR = 3.01, 95% CI = 1.53-5.92; p = 0.0017), TA or TT in rs822337 (OR = 2.45, 95% CI = 1.22-4.93; p = 0.0098), and CT or CC of rs822338 (OR = 2.23, 95% CI = 1.05-4.73; p = 0.028) were at an increased risk of developing invasive melanomas. Cases with the AG or GG genotype in rs2297136 presented a lower risk (OR = 0.29, 95% CI = 0.11-0.75; p = 0.0038) of invasive MM. The genetic analysis at the haplotype level resulted in similar findings (OR: 2.95, 95% CI: 1.08-8.10), p = 0.036). Furthermore, patients carrying the homozygous AA genotype in rs2297136 had thicker tumours than those harbouring the AG or GG (1.4 mm vs. 1.0 and 0.8 mm; p = 0.030). No significant association was found between the studied SNPs and melanoma-specific survival (MSS) nor progression-free survival (PFS). Conclusions: Current results suggest that SNPs rs822336, rs822337, rs822338, and rs2297136 genotypes in the PDCDL1 gene are associated with the risk of tumour invasiveness and tumour thickness in MM. Further studies on SNPs considering genetic and epigenetic factors are needed for a better understanding of malignant melanoma susceptibility and its prognosis.
Collapse
Affiliation(s)
- Elizabeth Córdoba-Lanús
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, Avda. Astrofísico Sánchez, s/n, 38296 San Cristóbal de La Laguna, Spain; (O.G.-P.)
- Consorcio Centro de Investigación Biomédica (CIBER) de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Omar García-Pérez
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, Avda. Astrofísico Sánchez, s/n, 38296 San Cristóbal de La Laguna, Spain; (O.G.-P.)
- Consorcio Centro de Investigación Biomédica (CIBER) de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Leticia Melgar-Vilaplana
- Pathology Department, Hospital Universitario Nuestra Señora de Candelaria, Ctra. Gral. del Rosario, 145, 38010 Santa Cruz de Tenerife, Spain;
| | - Angélica Domínguez-de-Barros
- Instituto Universitario de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), Universidad de La Laguna, Avda. Astrofísico Sánchez, s/n, 38296 San Cristóbal de La Laguna, Spain; (O.G.-P.)
- Consorcio Centro de Investigación Biomédica (CIBER) de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ricardo Fernández-de-Misa
- Research Unit, Hospital Universitario Nuestra Señora de Candelaria, Ctra. Gral. del Rosario, 145, 38010 Santa Cruz de Tenerife, Spain
- Dermatology Department, Hospital Universitario Nuestra Señora de Candelaria, Ctra. Gral. del Rosario, 145, 38010 Santa Cruz de Tenerife, Spain
- Department of Internal Medicine, Dermatology and Psychiatry, Universidad de La Laguna, 38071 San Cristóbal de La Laguna, Spain
| |
Collapse
|
18
|
Shen Y, Connolly E, Aiello M, Zhou C, Chappa P, Song H, Tippitak P, Clark T, Cardenas M, Prokhnevska N, Mariniello A, De Bruyker I, Pagadala MS, Dhere VR, Rafiq S, Kesarwala AH, Orthwein A, Thomas SN, Zhang SL, Khan MK, Dixon JB, Lesinski GB, Lowe MC, Kissick H, Yu DS, Paulos CM, Schmitt NC, Buchwald ZS. Combination radiation and αPD-L1 enhance tumor control by stimulating CD8+ PD-1+ TCF-1+ T cells in the tumor-draining lymph node. Nat Commun 2025; 16:3522. [PMID: 40229241 PMCID: PMC11997041 DOI: 10.1038/s41467-025-58510-1] [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: 02/12/2024] [Accepted: 03/19/2025] [Indexed: 04/16/2025] Open
Abstract
Combination radiotherapy (RT) and αPD-L1 therapy has potential to enhance local and distant (abscopal) tumor control, however, clinical results in humans have been variable. Using murine melanoma models, we found RT + αPD-L1 increases intra-tumor progenitor CD8+ PD-1+ TCF-1+ T cells. This increase depends on trafficking of the PD-1+ TCF-1+ cells from the tumor-draining lymph node (TdLN) to the tumor. RT alone promotes the expansion and differentiation of the TdLN derived PD-1+ TCF-1+ cells into TIM-3+ GZMB+ TCF-1- effector-like cells in the tumor with further enhancement after the addition of αPD-L1. In the TdLN, combination therapy enriches for a novel PD-1+ TCF-1+ TOX- LY6A+ subset with expression of a type I interferon and migratory signature. This subset is able to traffic to the tumor and differentiate into TIM-3+ TCF-1- cells. Finally, we found that ablation of the PD-1+ TCF-1+ T cell population attenuates the enhanced tumor control observed with combination RT + αPD-L1. These results suggest that abscopal response failures may be secondary to impaired stimulation of TdLN CD8+ PD-1 + TCF-1+ T cells or an inability of PD-1+ TCF-1+ cells in the TdLN to traffic to the tumor.
Collapse
Affiliation(s)
- Yang Shen
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Erin Connolly
- Bioinformatics Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Meili Aiello
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Chengjing Zhou
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Prasanthi Chappa
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Haorui Song
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Patan Tippitak
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Tarralyn Clark
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Maria Cardenas
- Department of Urology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Nataliya Prokhnevska
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai (ICMMS), New York City, NY, USA
| | - Annapaola Mariniello
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Isabelle De Bruyker
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Meghana S Pagadala
- Medical Scientist Training Program, University of California San Diego, La Jolla, CA, USA
| | - Vishal R Dhere
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sarwish Rafiq
- Department of Hematology and Medical Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Aparna H Kesarwala
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Alexandre Orthwein
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Susan N Thomas
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Shirley L Zhang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mohammad K Khan
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - J Brandon Dixon
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Gregory B Lesinski
- Department of Hematology and Medical Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Michael C Lowe
- Department of Surgery and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Haydn Kissick
- Department of Urology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - David S Yu
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Chrystal M Paulos
- Department of Surgery and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Nicole C Schmitt
- Department of Otolaryngology - Head and Neck Surgery and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Zachary S Buchwald
- Department of Radiation Oncology and Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| |
Collapse
|
19
|
Wei J, Li D, Long H, Han M. Immune microenvironment of tumor-draining lymph nodes: insights for immunotherapy. Front Immunol 2025; 16:1562797. [PMID: 40292299 PMCID: PMC12021902 DOI: 10.3389/fimmu.2025.1562797] [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: 01/18/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Tumor-draining lymph nodes (TDLNs) play a crucial role in modulating tumor immune responses and influencing the efficacy of immunotherapy. However, our current understanding of the microenvironment within these lymph nodes remains limited. Tumors not only impair the anti-tumor activity of CD8+ T cells by creating an immunosuppressive microenvironment, but they also facilitate immune evasion and promote metastasis by altering the structure and function of TDLNs. Research has shown that tumor-specific memory CD8+ T cells (TTSM) within TDLNs are essential for the efficacy of immune checkpoint inhibitors, such as PD-1/PD-L1 blockers. Moreover, the abnormal structure of TDLNs, along with the presence of immunosuppressive cells-such as regulatory T cells (Tregs), regulatory B cells (Bregs), and immunosuppressive dendritic cells (DCs)-contributes to tumor-mediated immune evasion. Therefore, gaining a deeper understanding of the immune microenvironment within TDLNs is essential for improving the effectiveness of immunotherapies and developing novel therapeutic strategies. This review explores various TDLN-based therapeutic strategies, addressing the controversies surrounding lymph node dissection, the use of TDLNs as a source of tumor-infiltrating lymphocytes (TILs) for therapy, targeting immunosuppressive cells within TDLNs, and methods to reverse the structural abnormalities of TDLNs. These strategies offer valuable insights and potential directions for advancing tumor immunotherapy.
Collapse
Affiliation(s)
- Jiahuan Wei
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Daozhang Li
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Haixia Long
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Mei Han
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| |
Collapse
|
20
|
Arai M, Tanaka N, Takamatsu K, Murakami T, Mikami S, Imamura T, Nakamura K, Nishihara H, Oya M. Prognostic impact and landscape of cellular CXCR5 chemokine receptor expression in clear-cell renal cell carcinoma. Cancer Immunol Immunother 2025; 74:166. [PMID: 40208344 PMCID: PMC11985720 DOI: 10.1007/s00262-025-04020-4] [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: 11/04/2024] [Accepted: 03/13/2025] [Indexed: 04/11/2025]
Abstract
CXCR5 is a chemokine receptor that promotes B cell follicular formation and antibody production. Indeed, CXCR5 has been found to be expressed in a variety of cancers; however, the role of CXCR5 expression in clear-cell renal cell carcinoma (ccRCC) remains unclear. We aimed to determine the impact of cellular CXCR5 expression on cancer outcomes, the PD-1/PD-L1 axis, and genetic states in patients with ccRCC. First, multiplex immunofluorescence staining for CXCR5, CD4, CD8, and AE1/AE3, along with automated single-cell counting, was performed to assess cellular CXCR5 expression in ccRCC and its association with prognosis. Second, the tumour microenvironment (TME) was analysed, with a focus on the relationship between the PD-1/PD-L1 axis and CXCR5 expression. Finally, an integrated analysis of CXCR5 expression and genomic mutation information was conducted to reveal the genetic background underlying CXCR5 expression. A total of 105 ccRCC patients were included. Among the 696,964 cells analysed, the distribution of CXCR5-expressing cells was as follows: 30% CXCR5+CD4+ cells, 9% CXCR5+CD8+ cells, and 26% CXCR5+AE1/AE3+ cells. Survival analysis revealed that tumours with low-CXCR5+CD8+ cells had a poor prognosis; TME analysis revealed a relationship between low-CXCR5+CD8+ status and a highly suppressive PD-L1-positive immune environment. Genomic analysis revealed a correlation between low-CXCR5+CD8+ status and high rates of alterations in chromatin remodelling genes, including PBRM1. This study highlights the significance of CXCR5+CD8+ cells in ccRCC, demonstrating their clinical implications and revealing the immunogenomic landscape underlying CXCR5 expression.
Collapse
Affiliation(s)
- Masashi Arai
- Department of Urology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Nobuyuki Tanaka
- Department of Urology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| | - Kimiharu Takamatsu
- Department of Urology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Tetsushi Murakami
- Department of Urology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Shuji Mikami
- Department of Diagnostic Pathology, Keio University Hospital, Tokyo, Japan
- Department of Diagnostic Pathology, National Hospital Organization Saitama Hospital, Wako, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Graduate School of Medicine, Ehime University, Toon, Japan
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| |
Collapse
|
21
|
Ahn SY, Ho TL, Ko EJ. Evaluation of the influenza vaccine protection in the house dust mite-induced chronic allergic asthma mice model and the evaluation of squalene oil in water emulsion as an adjuvant candidate. Respir Res 2025; 26:132. [PMID: 40205548 PMCID: PMC11984255 DOI: 10.1186/s12931-025-03209-6] [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: 10/04/2024] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Despite the importance of influenza vaccination in asthma patients, the efficacy of this vaccine in asthma has not been well elucidated. We aimed to compare the efficacy of an influenza vaccine of the asthmatic and control mice. We also evaluated the efficacy of AddaVax™ as an adjuvant candidate, which is equivalent to the MF59 influenza vaccine adjuvant in the elderly. METHOD House dust mite extracts were intranasally injected into six-week-old female BALB/c mice to induce chronic allergic asthma. Antibody responses after split-inactivated A/Puerto Rico/8/34 H1N1 influenza vaccination with or without AddaVax™ adjuvant were measured using ELISA. Homologous viral protection was determined by measuring the survival rate, lung inflammation level, and lung virus titer after challenge with the human influenza virus strain A/Puerto Rico/8/1934 H1N1. Antigen-specific T cell responses were determined using flow cytometry. RESULT The chronic asthma mice immunized with split-inactivated A/Puerto Rico/8/34 H1N1 influenza vaccine showed significant weight loss and higher lung viral load after homologous influenza infection than naïve vaccinated mice. Antigen-specific IgG, IgG1, and IgG2a production did not differ between the naïve and asthma mice. However, serum HI titer was lower in asthma-vaccinated mice after infection. The application of AddaVax™ to a vaccine for mice with asthma enhanced the efficacy of homologous antiviral protection but elicited eosinophil infiltration in the lungs after homologous influenza virus infection. CONCLUSION The immune response after split inactivated A/PR8 vaccine differed between asthma and naïve mice, particularly in terms of antibody activity and T cell populations. This study enhances our understanding of how asthma status may influence the effectiveness of influenza vaccine and offers insights into the AddaVax™-induced eosinophilic inflammation, guiding the development of virus vaccine strategies for both healthy individuals and asthma patients.
Collapse
Affiliation(s)
- So Yeon Ahn
- Department of Veterinary Medicine and Veterinary Medical Research Institute, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Thi Len Ho
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, 63243, Republic of Korea
| | - Eun-Ju Ko
- Department of Veterinary Medicine and Veterinary Medical Research Institute, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea.
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, 63243, Republic of Korea.
- Bio-Health Materials Core-Facility Center, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|
22
|
Moës B, Krueger J, Kazanova A, Liu C, Gao Y, Ponnoor NA, Castoun-Puckett L, Lazo ACO, Huong L, Cabald AL, Tu TH, Rudd CE. GSK-3 regulates CD4-CD8 cooperation needed to generate super-armed CD8+ cytolytic T cells against tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.08.642085. [PMID: 40161618 PMCID: PMC11952298 DOI: 10.1101/2025.03.08.642085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
While immune checkpoint blockade (ICB) has revolutionized cancer treatment, the key T-cell signaling pathways responsible for its potency remain unclear. GSK-3 is an inhibitory kinase that is most active in resting T-cells. In this study, we demonstrate that GSK-3 facilitates PD-1 blockade, an effect seen by modulating CD4 T-cell help for CD8+ CTL responses against ICB resistant tumors. We show that GSK-3 controls metabolic reprogramming towards glycolysis and synergizes with PD-1 to induce a transcriptional program that reduces suppressive CD4+ Treg numbers while generating super-armed effector-memory CD8+ CTLs that express an unprecedented 7/9 granzymes from the genome. Crucially, we found that GSK-3 cooperates with PD-1 blockade to determine the dependency of CD8+ CTLs on help from CD4+ T-cells. Our study unravels a novel cooperative PD-1 blockade-dependent signaling pathway that potentiates CTL responses against tumors, offering a new strategy to overcome immunotherapy resistance by modulating CD4+ helper and CD8+ cytotoxic functions. Significance This study demonstrates for the first time that GSK-3 controls the crosstalk between CD4+ and CD8+ T cells, synergizing with anti-PD-1 therapy to overcome resistance to checkpoint blockade and to generate super-armed CD8+ effector cells in cancer immunotherapy. This newly uncovered GSK-3-dependent CD4-CD8 T-cell crosstalk mechanism presents a new approach to enhance anti-PD-1 immunotherapy.
Collapse
|
23
|
Santoni M, Rizzo A, Massari F. Unlocking the mechanisms underlying the activity of pembrolizumab plus enfortumab vedotin in patients with urothelial carcinoma. Expert Opin Investig Drugs 2025; 34:259-265. [PMID: 40012129 DOI: 10.1080/13543784.2025.2473695] [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: 09/03/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 02/28/2025]
Abstract
INTRODUCTION Urothelial carcinoma (UC) is frequently associated with a poor prognosis in patients with advanced disease. A strong biological rationale supports the investigation of combining antibody-drug conjugates (ADCs) with immunotherapy to overcome the occurrence of resistance and improve patient outcomes. AREAS COVERED In this review, we illustrate the mechanisms of action of pembrolizumab and enfortumab vedotin (EV) and the immune and biological rationales underlying their synergy in mUC patients. EXPERT OPINION The results of the combination of EV and pembrolizumab represent a ray of light in the therapeutic scenario of mUC patients. A deeper understanding of the mechanisms underlying the synergistic effects of these agents will be crucial to reduce drug-resistance and further improve the outcome of mUC patients.
Collapse
Affiliation(s)
- Matteo Santoni
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, Macerata, Italy
| | - Alessandro Rizzo
- Struttura S.S.D.C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori "Giovanni Paolo II, Bari, Italy
| | - Francesco Massari
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy, Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
24
|
Chu T, Wu M, Hoellbacher B, de Almeida GP, Wurmser C, Berner J, Donhauser LV, Gerullis AK, Lin S, Cepeda-Mayorga JD, Kilb II, Bongers L, Toppeta F, Strobl P, Youngblood B, Schulz AM, Zippelius A, Knolle PA, Heinig M, Hackstein CP, Zehn D. Precursors of exhausted T cells are pre-emptively formed in acute infection. Nature 2025; 640:782-792. [PMID: 39778709 PMCID: PMC12003159 DOI: 10.1038/s41586-024-08451-4] [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: 02/10/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025]
Abstract
T cell exhaustion limits effector T cell function in chronic infection and tumours1,2. The development of these hypofunctional T cells and of their precursors was considered to require stimulatory conditions that are met only after persistent exposure to antigen and inflammation. Here we show, however, that similar T cell populations exist in the early phase of acute infections1,2. At that stage, the early developing TCF1+ precursor population exhibits an unexpected diversity; it includes precursors of normal memory T cells, but also cells with phenotypic, gene-expression and epigenetic profiles that resemble those of precursors of exhausted T cells found in chronic infections. We show that high ligand affinity promotes and PD-1 signalling restricts the development of these precursors. Although the exhausted precursors are at first found frequently, they decline without being completely lost in infections that the immune system resolves. We therefore conclude that precursor T cells with at least two distinct phenotypes are pre-emptively generated irrespective of the outcome of an infection.
Collapse
Affiliation(s)
- Talyn Chu
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Cancer Immunology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Ming Wu
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
| | - Barbara Hoellbacher
- Institute of Computational Biology, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany
- Department of Informatics, Technical University of Munich, Garching, Germany
| | - Gustavo P de Almeida
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Christine Wurmser
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Jacqueline Berner
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Lara V Donhauser
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Ann-Katrin Gerullis
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Siran Lin
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - J Diego Cepeda-Mayorga
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Iman I Kilb
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Lukas Bongers
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Fabio Toppeta
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Philipp Strobl
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Ben Youngblood
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Anna M Schulz
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Alfred Zippelius
- Cancer Immunology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Percy A Knolle
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Matthias Heinig
- Institute of Computational Biology, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt, Neuherberg, Germany.
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany.
| | - Carl-Philipp Hackstein
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Institute of Molecular Immunology, School of Medicine and Health, Technical University of Munich, Munich, Germany.
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
| |
Collapse
|
25
|
Rausch L, Kallies A. Molecular Mechanisms Governing CD8 T Cell Differentiation and Checkpoint Inhibitor Response in Cancer. Annu Rev Immunol 2025; 43:515-543. [PMID: 40279308 DOI: 10.1146/annurev-immunol-082223-044122] [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] [Indexed: 04/27/2025]
Abstract
CD8 T cells play a critical role in antitumor immunity. However, over time, they often become dysfunctional or exhausted and ultimately fail to control tumor growth. To effectively harness CD8 T cells for cancer immunotherapy, a detailed understanding of the mechanisms that govern their differentiation and function is crucial. This review summarizes our current knowledge of the molecular pathways that regulate CD8 T cell heterogeneity and function in chronic infection and cancer and outlines how T cells respond to therapeutic checkpoint blockade. We explore how T cell-intrinsic and -extrinsic factors influence CD8 T cell differentiation, fate choices, and functional states and ultimately dictate their response to therapy. Identifying cells that orchestrate long-term antitumor immunity and understanding the mechanisms that govern their development and persistence are critical steps toward improving cancer immunotherapy.
Collapse
Affiliation(s)
- Lisa Rausch
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia;
| | - Axel Kallies
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia;
| |
Collapse
|
26
|
McManus DT, Valanparambil RM, Medina CB, Scharer CD, McGuire DJ, Sobierajska E, Hu Y, Chang DY, Wieland A, Lee J, Nasti TH, Hashimoto M, Ross JL, Prokhnevska N, Cardenas MA, Gill AL, Clark EC, Abadie K, Kumar AJ, Kaye J, Au-Yeung BB, Kueh HY, Kissick HT, Ahmed R. An early precursor CD8 + T cell that adapts to acute or chronic viral infection. Nature 2025; 640:772-781. [PMID: 39778710 DOI: 10.1038/s41586-024-08562-y] [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: 02/02/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025]
Abstract
This study examines the origin and differentiation of stem-like CD8+ T cells that are essential for sustained T cell immunity in chronic viral infections and cancer and also have a key role in PD-1 directed immunotherapy1-10. These PD-1+TCF-1+TOX+ stem-like CD8+ T cells (also known as precursors of exhausted T cells8,9) have a distinct program that enables them to adapt to chronic antigen stimulation. Here, using the mouse model of chronic lymphocytic choriomeningitis virus (LCMV) infection, we find that virus-specific stem-like CD8+ T cells are generated early (day 5) during chronic infection, suggesting that this crucial fate commitment occurs irrespective of the infection outcome. Indeed, we find that nearly identical populations of stem-like CD8+ T cells were generated early during acute or chronic LCMV infection, and that antigen was essential for maintaining the stem-like phenotype. We performed reciprocal adoptive transfer experiments to determine the fate of these early stem-like CD8+ T cells after viral clearance versus persistence. After transfer of day 5 stem-like CD8+ T cells from chronically infected mice into acutely infected mice, these cells downregulated canonical markers of the chronic stem-like CD8+ T cells and expressed markers (CD127 and CD62L) associated with central memory CD8+ T cells. Reciprocally, when day 5 stem-like cells from acutely infected mice were transferred into chronically infected mice, these CD8+ T cells functioned like chronic resource cells and responded effectively to PD-1 therapy. These findings highlight the ability of these early PD-1+TCF-1+TOX+ stem-like CD8+ T cells to adapt their differentiation trajectory to either an acute or a chronic viral infection. Importantly, our study shows that the host is prepared a priori to deal with a potential chronic infection.
Collapse
Affiliation(s)
- Daniel T McManus
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rajesh M Valanparambil
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher B Medina
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Christopher D Scharer
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Donald J McGuire
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Ewelina Sobierajska
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yinghong Hu
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Daniel Y Chang
- Department of Pathology, Mass General Brigham, Harvard Medical School, Boston, MA, USA
| | - Andreas Wieland
- Department of Otolaryngology, The Ohio State University College of Medicine, Columbus, OH, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-The James, Columbus, OH, USA
| | - Judong Lee
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Tahseen H Nasti
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Masao Hashimoto
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - James L Ross
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Nataliya Prokhnevska
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria A Cardenas
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Amanda L Gill
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
| | - Elisa C Clark
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Kathleen Abadie
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Arjun J Kumar
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jonathan Kaye
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Byron B Au-Yeung
- Division of Immunology, Lowance Center for Human Immunology, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Hao Yuan Kueh
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Haydn T Kissick
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA.
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, USA.
- Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| |
Collapse
|
27
|
Nair R, Somasundaram V, Kuriakose A, Krishn SR, Raben D, Salazar R, Nair P. Deciphering T-cell exhaustion in the tumor microenvironment: paving the way for innovative solid tumor therapies. Front Immunol 2025; 16:1548234. [PMID: 40236693 PMCID: PMC11996672 DOI: 10.3389/fimmu.2025.1548234] [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: 12/19/2024] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
In solid tumors, the tumor microenvironment (TME) is a complex mix of tumor, immune, stromal cells, fibroblasts, and the extracellular matrix. Cytotoxic T lymphocytes (CTLs) constitute a fraction of immune cells that may infiltrate into the TME. The primary function of these T-cells is to detect and eliminate tumor cells. However, due to the immunosuppressive factors present in the TME primarily mediated by Myeloid-Derived Suppressor Cells (MDSCs), Tumor associated macrophages (TAMs), Cancer Associated Fibroblasts (CAFs) as well as the tumor cells themselves, T-cells fail to differentiate into effector cells or become dysfunctional and are unable to eliminate the tumor. In addition, chronic antigen stimulation within the TME also leads to a phenomenon, first identified in chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, where the T-cells become exhausted and lose their effector functions. Exhausted T-cells (Tex) are characterized by the presence of remarkably conserved inhibitory receptors, transcription and signaling factors and the downregulation of key effector molecules. Tex cells have been identified in various malignancies, including melanoma, colorectal and hepatocellular cancers. Recent studies have indicated novel strategies to reverse T-cell exhaustion. These include checkpoint inhibitor blockade targeting programmed cell death protein 1 (PD-1), T-cell immunoglobulin and mucin-domain containing-3 (Tim-3), cytotoxic T-lymphocyte associated protein 4 (CTLA-4), or combinations of different immune checkpoint therapies (ICTs) or combination of ICTs with cytokine co-stimulation. In this review, we discuss aspects of T-cell dysfunction within the TME with a focus on T-cell exhaustion. We believe that gaining insight into the mechanisms of T-cell exhaustion within the TME of human solid tumors will pave the way for developing therapeutic strategies to target and potentially re-invigorate exhausted T-cells in cancer.
Collapse
Affiliation(s)
- Reshmi Nair
- Syngene International Limited, Bengaluru, India
| | | | | | | | - David Raben
- Bicara Therapeutics, Boston, MA, United States
| | | | - Pradip Nair
- Syngene International Limited, Bengaluru, India
| |
Collapse
|
28
|
Aksak-Wąs B, Skonieczna-Żydecka K, Parczewski M, Hrynkiewicz R, Lewandowski F, Serwin K, Mielczak K, Lenkiewicz F, Niedźwiedzka-Rystwej P. Rethinking HIV treatment: How non-integrase strand regimens may hold the key to better immune health. HIV Med 2025. [PMID: 40156175 DOI: 10.1111/hiv.70020] [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: 11/24/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
PURPOSE HIV outcome changed drastically with antiretroviral (ARV) therapy, especially after the introduction of second-generation integrase strand transfer inhibitors (INSTIs). Despite these advances, however, chronic immune activation and exhaustion, marked by programmed cell death 1 (PD-1) and programmed death ligand 1 (PD-L1) upregulation, persist in patients. This study investigates the impact of various ARV regimens on these immune exhaustion markers in newly diagnosed HIV patients over 12 months, taking into consideration cardiovascular risk. METHODS This study included 58 newly diagnosed patients with HIV at Pomeranian Medical University, Szczecin, Poland. Participants received ARV regimens classified as INSTI + tenofovir alafenamide, INSTI + tenofovir disoproxil fumarate, or non-INSTI-based (VARIA). Flow cytometry was used to assess PD-1 and PD-L1 expression on CD3+, CD3+CD4+, CD3+CD8+ and CD19+ lymphocytes. Statistical analyses included Wilcoxon paired tests, Kruskal-Wallis tests and multivariate regression, with validation through residual analysis and linear discriminant analysis (LDA). RESULTS INSTI-based regimens were linked to higher PD-1 expression on CD3+ and CD3+CD4+ lymphocytes, indicating increased immune exhaustion. Conversely, non-INSTI regimens were associated with lower PD-1 levels, suggesting better retention of immune function. A positive correlation between cardiovascular risk a prediction model to estimate 10-year fatal and non-fatal cardiovascular disease (SCORE2) and PD-1 expression was observed. However, the modest explanatory power of the models suggests variability in the effects of different ARV regimens. CONCLUSION This study challenges the assumption that INSTI-based ARV regimens are universally superior, suggesting that non-INSTI therapies may better preserve immune function by reducing PD-1 expression. These findings highlight the potential benefits of non-INSTI regimens in improving long-term clinical outcomes in HIV treatment, warranting further research.
Collapse
Affiliation(s)
- Bogusz Aksak-Wąs
- Department of Infectious, Tropical Diseases and Acquired Immunodeficiency, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | | - Miłosz Parczewski
- Department of Infectious, Tropical Diseases and Acquired Immunodeficiency, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Rafał Hrynkiewicz
- Institute of Biology, University of Szczecin, Szczecin, Poland
- Center for Experimental Immunology and Immunobiology in Infectious Diseases and Cancer, University of Szczecin, Szczecin, Poland
| | - Filip Lewandowski
- Institute of Biology, University of Szczecin, Szczecin, Poland
- Center for Experimental Immunology and Immunobiology in Infectious Diseases and Cancer, University of Szczecin, Szczecin, Poland
| | - Karol Serwin
- Department of Infectious, Tropical Diseases and Acquired Immunodeficiency, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Kaja Mielczak
- Department of Infectious, Tropical Diseases and Acquired Immunodeficiency, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Franciszek Lenkiewicz
- Department of Infectious, Tropical Diseases and Acquired Immunodeficiency, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Paulina Niedźwiedzka-Rystwej
- Institute of Biology, University of Szczecin, Szczecin, Poland
- Center for Experimental Immunology and Immunobiology in Infectious Diseases and Cancer, University of Szczecin, Szczecin, Poland
| |
Collapse
|
29
|
Viramontes KM, Thone MN, De La Torre JJ, Neubert EN, DeRogatis JM, Garcia C, Henriquez ML, Tinoco R. Contrasting roles of PSGL-1 and PD-1 in regulating T-cell exhaustion and function during chronic viral infection. J Virol 2025; 99:e0224224. [PMID: 39912665 PMCID: PMC11915808 DOI: 10.1128/jvi.02242-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/19/2025] [Indexed: 02/07/2025] Open
Abstract
Immune checkpoints are critical regulators of T-cell exhaustion, impairing their ability to eliminate antigens present during chronic viral infections. Current immune checkpoint inhibitors (ICIs) used in the clinic aim to reinvigorate exhausted T cells; yet, most patients fail to respond or develop resistance to these therapies, underscoring the need to better understand these immunosuppressive pathways. PSGL-1 (Selplg), a recently discovered immune checkpoint, negatively regulates T-cell function. We investigated the cell-intrinsic effects of PSGL-1, PD-1, and combined deletion on CD8+ T cells during chronic viral infection. We found that combined PSGL-1 and PD-1 (Selplg-/-Pdcd1-/-) deficiency in CD8+ T cells increased their frequencies and numbers throughout chronic infection compared to the wild type. This phenotype was primarily driven by PD-1 deficiency. Furthermore, while PD-1 deletion increased virus-specific T-cell frequencies, it was detrimental to their function. Conversely, PSGL-1 deletion improved T-cell function but resulted in lower frequencies and numbers. The primary mechanism behind these differences in cell maintenance was driven by proliferation rather than survival. Combined PSGL-1 and PD-1 deletion resulted in defective T-cell differentiation, driving cells from a progenitor self-renewal state to a more terminal dysfunctional state. These findings suggest that PD-1 and PSGL-1 have distinct, yet complementary, roles in regulating T-cell exhaustion and differentiation during chronic viral infection. Overall, this study provides novel insights into the individual and combined roles of PSGL-1 and PD-1 in CD8+ T-cell exhaustion. It underscores the potential of targeting these checkpoints in a more dynamic and sequential manner to optimize virus-specific T-cell responses, offering critical perspectives for improving therapeutic strategies aimed at reinvigorating exhausted CD8+ T cells.IMPORTANCEOur findings provide a comprehensive analysis of how the dual deletion of PD-1 and PSGL-1 impacts the response and function of virus-specific CD8+ T cells, revealing novel insights into their roles in chronic infection. Notably, our findings show that while PD-1 deletion enhances T-cell frequencies, it paradoxically reduces T-cell functionality. Conversely, PSGL-1 deletion improves T-cell function but reduces their survival. Whereas the combined deletion of PSGL-1 and PD-1 in CD8+ T cells improved their survival but decreased their function and progenitor-exhausted phenotypes during infection. We believe our study advances the understanding of immune checkpoint regulation in chronic infections and has significant implications for developing more effective immune checkpoint inhibitor (ICI) therapies.
Collapse
Affiliation(s)
- Karla M. Viramontes
- Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California Irvine, Irvine, California, USA
| | - Melissa N. Thone
- Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California Irvine, Irvine, California, USA
| | - Jamie-Jean De La Torre
- Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California Irvine, Irvine, California, USA
| | - Emily N. Neubert
- Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California Irvine, Irvine, California, USA
| | - Julia M. DeRogatis
- Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California Irvine, Irvine, California, USA
| | - Chris Garcia
- Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California Irvine, Irvine, California, USA
| | - Monique L. Henriquez
- Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California Irvine, Irvine, California, USA
| | - Roberto Tinoco
- Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California Irvine, Irvine, California, USA
- Center for Virus Research, University of California Irvine, Irvine, California, USA
- Institute for Immunology, University of California Irvine, Irvine, California, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, California, USA
| |
Collapse
|
30
|
Xu Q, Li L, Zhu R. T Cell Exhaustion in Allergic Diseases and Allergen Immunotherapy: A Novel Biomarker? Curr Allergy Asthma Rep 2025; 25:18. [PMID: 40091122 DOI: 10.1007/s11882-025-01199-5] [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] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE OF REVIEW This review explores the emerging role of T cell exhaustion in allergic diseases and allergen immunotherapy (AIT). It aims to synthesize current knowledge on the mechanisms of T cell exhaustion, evaluate its potential involvement in allergic inflammation, and assess its implications as a novel biomarker for predicting and monitoring AIT efficacy. RECENT FINDINGS Recent studies highlight that T cell exhaustion, characterized by co-expression of inhibitory receptors (e.g., PD-1, CTLA-4, TIM-3), diminished cytokine production, and altered transcriptional profiles, may suppress type 2 inflammation in allergic diseases. In allergic asthma, exhausted CD4 + T cells exhibit upregulated inhibitory receptors, correlating with reduced IgE levels and airway hyperreactivity. During AIT, prolonged high-dose allergen exposure drives allergen-specific Th2 and T follicular helper (Tfh) cell exhaustion, potentially contributing to immune tolerance. Notably, clinical improvements in AIT correlate with depletion of allergen-specific Th2 cells and persistent expression of exhaustion markers (e.g., PD-1, CTLA-4) during maintenance phases. Blockade of inhibitory receptors (e.g., PD-1) enhances T cell activation, underscoring their dual regulatory role in allergy. T cell exhaustion represents a double-edged sword in allergy: it may dampen pathological inflammation in allergic diseases while serving as a mechanism for AIT-induced tolerance. The co-expression of inhibitory receptors on allergen-specific T cells emerges as a promising biomarker for AIT efficacy. Future research should clarify the transcriptional and metabolic drivers of exhaustion in allergy, validate its role across diverse allergic conditions, and optimize strategies to harness T cell exhaustion for durable immune tolerance. These insights could revolutionize therapeutic approaches and biomarker development in allergy management.
Collapse
Affiliation(s)
- Qingxiu Xu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Le Li
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rongfei Zhu
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
31
|
Lin Y, Song Y, Zhang Y, Li X, Kan L, Han S. New insights on anti-tumor immunity of CD8 + T cells: cancer stem cells, tumor immune microenvironment and immunotherapy. J Transl Med 2025; 23:341. [PMID: 40097979 PMCID: PMC11912710 DOI: 10.1186/s12967-025-06291-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/23/2025] [Indexed: 03/19/2025] Open
Abstract
Recent breakthroughs in tumor immunotherapy have confirmed the capacity of the immune system to fight several cancers. The effective means of treating cancer involves accelerating the death of tumor cells and improving patient immunity. Dynamic changes in the tumor immune microenvironment alter the actual effects of anti-tumor drug production and may trigger favorable or unfavorable immune responses by modulating tumor-infiltrating lymphocytes. Notably, CD8+ T cells are one of the primary tumor-infiltrating immune cells that provide anti-tumor response. Tumor cells and tumor stem cells will resist or evade destruction through various mechanisms as CD8+ T cells exert their anti-tumor function. This paper reviews the research on the regulation of tumor development and prognosis by cancer stem cells that directly or indirectly alter the role of tumor-infiltrating CD8+ T cells. We also discuss related immunotherapy strategies.
Collapse
Affiliation(s)
- Yibin Lin
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yifu Song
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yaochuan Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaodong Li
- Department of Neurosurgery, Siping Central People's Hospital, Siping, Jilin, 136000, China
| | - Liang Kan
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Sheng Han
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
32
|
Yoshinaga K, Hirano T, Umemoto S, Kadowaki Y, Matsunaga T, Suzuki M. Effect of Anti-Programmed Cell Death-1 Antibody on Middle Ear Mucosal Immune Response to Intranasal Administration of Haemophilus influenzae Outer Membrane Protein. Vaccines (Basel) 2025; 13:313. [PMID: 40266194 PMCID: PMC11946078 DOI: 10.3390/vaccines13030313] [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: 01/24/2025] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025] Open
Abstract
Background/Objectives: Acute otitis media is a common pediatric infection caused primarily by nontypeable Haemophilus influenzae. With rising antibiotic resistance, vaccines are essential for combating this public health issue. Although the PD-1/PD-L1 pathway has been extensively studied for its role in tumor immunity, its impact on mucosal immunity, particularly in vaccine responses, is unclear. Methods: BALB/c mice were intranasally immunized with nontypeable H. influenzae outer membrane protein and treated with anti-PD-L1 antibodies. Immune responses were evaluated in middle ear mucosa (MEM), the cervical lymph node, and the spleen using an enzyme-linked immunosorbent assay, an enzyme-linked immunospot assay, and flow cytometry. The effects on CD4+ T cells, T follicular helper (Tfh) cells, and B-cell differentiation were analyzed. Results: Anti-PD-L1 antibody treatment increased CD3+CD4+CD185+ (CXCR5+) Tfh cells in MEM, which play a crucial role in supporting B-cell activation and antibody production. This correlated with a significant increase in IgA- and IgG-producing cells in MEM, which enhanced local bacterial clearance. Although B-cell activation and differentiation into plasmablasts were observed in MEM, no significant changes were noted in the cervical lymph node and spleen, suggesting a localized enhancement of mucosal immunity. Conclusions: Anti-PD-L1 antibodies promoted Tfh cell expansion and B-cell differentiation in MEM, leading to enhanced antibody production and improved bacterial clearance. These findings suggest that PD-L1 blockade can potentiate mucosal vaccine-induced immunity by strengthening local humoral responses. This supports its potential application in developing intranasal vaccines for acute otitis media.
Collapse
Affiliation(s)
| | - Takashi Hirano
- Department of Otorhinolaryngology& Head and Neck Surgery, Faculty of Medicine, Oita University, Oita 879-5593, Japan; (K.Y.)
| | | | | | | | | |
Collapse
|
33
|
Chen HZ, Kim NH, Nishizaki D, Nesline MK, Conroy JM, DePietro P, Pabla S, Kato S, Kurzrock R. PD-1 transcriptomic landscape across cancers and implications for immune checkpoint blockade outcome. NPJ Genom Med 2025; 10:21. [PMID: 40069238 PMCID: PMC11897377 DOI: 10.1038/s41525-025-00465-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/20/2025] [Indexed: 03/15/2025] Open
Abstract
Programmed cell death protein 1 (PD-1) is a critical immune checkpoint receptor and a target for cancer immune checkpoint inhibitors (ICI). We investigated PD-1 transcript expression across cancer types and its correlations to clinical outcomes. Using a reference population, PD-1 expression was calculated as percentiles in 489 of 514 patients (31 cancer types) with advanced/metastatic disease. PD-1 RNA expression varied across and within cancer types; pancreatic and liver/bile duct malignancies displayed the highest rates of high PD-1 (21.82% and 21.05%, respectively). Elevated CTLA-4, LAG-3, and TIGIT RNA expression were independently correlated with high PD-1. Although high PD-1 was not associated with outcome in immunotherapy-naïve patients (n = 272), in patients who received ICIs (n = 217), high PD-1 transcript expression was independently correlated with prolonged survival (hazard ratio 0.40; 95%CI, 0.18-0.92). This study identifies PD-1 as an important biomarker in predicting ICI outcomes, and advocates for comprehensive immunogenomic profiling in cancer management.
Collapse
Affiliation(s)
- Hui-Zi Chen
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Na Hyun Kim
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.
| | - Daisuke Nishizaki
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | | | | | | | | | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.
- Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
34
|
Yang Y, Li X, Liu F, Ma M, Yang Y, Ruan C, Lu Y, Li X, Wang X, Shi Y, Zhang Z, Wang H, Cheng Z, Wu D. Immunometabolite L-2-HG promotes epigenetic modification of exhausted T cells and improves antitumor immunity. JCI Insight 2025; 10:e174600. [PMID: 40043713 PMCID: PMC11981629 DOI: 10.1172/jci.insight.174600] [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: 08/07/2023] [Accepted: 02/21/2025] [Indexed: 04/09/2025] Open
Abstract
This study aimed to explore the potential correlation between the metabolic intermediate L-2-hydroxyglutarate (L-2-HG) and T cell exhaustion, as well as the underlying mechanisms involved. In this study, we investigated the presence of exhausted T (Tex) cells in patients under certain conditions: HIV infection, chronic leukemia, and hepatocellular carcinoma. To gain insights into the epigenetic signatures and transcriptome changes in Tex cells, we employed a combination of RNA-seq and ATAC-seq analyses. To evaluate the impact of L-2-HG on mitochondrial function, differentiation, and antitumor capacity of Tex cells, we utilized in vitro cell culture experiments and animal tumor models. We observed mitochondrial depolarization and metabolic dysfunction in Tex cells, accompanied by a significant reduction in L-2-HG levels. Moreover, altered epigenetic characteristics were observed in Tex cells, including a substantial increase in H3K27me3 abundance. Culturing Tex cells with L-2-HG demonstrated improved mitochondrial metabolism, reduced H3K27me3 abundance, and enhanced memory T cell differentiation. In a mouse melanoma tumor model, L-2-HG-treated CD8+ T cells for adoptive therapy led to significantly reduced tumor volume and significantly enhanced effector function of T cells. The study revealed that L-2-HG acted as an immune metabolite through epigenetic modifications of Tex cells.
Collapse
Affiliation(s)
- Yanying Yang
- Department of Endocrinology, Zhongshan Hospital, and
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Endocrinology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaoyan Li
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangming Liu
- Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mingyue Ma
- Department of Endocrinology, Zhongshan Hospital, and
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Yang
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Chengchao Ruan
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yan Lu
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyang Li
- 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
| | - Xiangdong Wang
- Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yinghong Shi
- Liver Surgery Department of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhouli Cheng
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Duojiao Wu
- Shanghai Key Laboratory of Lung Inflammation and Injury, Zhongshan Hospital, Fudan University, Shanghai, China
- Jinshan Hospital Center for Tumor Diagnosis & Therapy, Jinshan Hospital, Fudan University, Shanghai, China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Xie L, Gong J, He Z, Zhang W, Wang H, Wu S, Wang X, Sun P, Cai L, Wu Z, Wang H. A Copper-Manganese Based Nanocomposite Induces Cuproptosis and Potentiates Anti-Tumor Immune Responses. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412174. [PMID: 39955646 DOI: 10.1002/smll.202412174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/26/2025] [Indexed: 02/17/2025]
Abstract
Cancer is one of the most important challenges worldwide with an increasing incidence. However, most of patients with malignant cancer receiving traditional therapies have tumor recurrence and short-term 5-year survival. Herein, a novel Cu2O-MnO@PEG (CMP) nanomaterial is developed to treat tumors. CMP directly mediates cuproptosis in tumor cells. Meanwhile, CMP potentiates anti-tumor immune responses in the tumor microenvironment (TME) to induce tumor regression. CMP improves the tumor antigen processing and presentation of dendritic cells and tumor-associated macrophages, and further promotes CD8+ T cell responses, especially for cytotoxic CD8+ T cells and transitory exhausted CD8+ T cells. Additionally, CMP downregulates the proportion of Treg cells and CTLA-4 expression on Treg cells. Notably, CMP induces systemic immune responses against distant tumors and long-term immune memory. Furthermore, CMP synergized with PD-L1 mAb promotes tumor inhibition and sustains the anti-tumor efficacy post PD-L1 mAb treatment. Collectively, this strategy has the clinically therapeutic potential for tumors by facilitating cuproptosis in tumor cells and anti-tumor immune responses.
Collapse
Affiliation(s)
- Luoyingzi Xie
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Jie Gong
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
- School of Clinical Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
- Department of Hepatobiliary Surgery, Leshan People's Hospital, Leshan, 614000, P. R. China
| | - Zhiqiang He
- Department of Dermatology, Southwest Hospital Jiangbei Area (The 958th hospital of Chinese People's Liberation Army), Chongqing, 400020, P. R. China
| | - Weinan Zhang
- Department of Urinary Nephropathy Center, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, P. R. China
| | - Haoyu Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
| | - Shitao Wu
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
- Graduate School of Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Xianxing Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
| | - Pijiang Sun
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
| | - Lei Cai
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
| | - Zhongjun Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Intelligent Medicine Engineering for Hepatopancreatobiliary Diseases, Chongqing, 401147, P. R. China
| |
Collapse
|
36
|
Özdemir BH, Baştürk B, Sayın CB, Haberal M. Programmed Death-Ligand 1 in Renal Allografts With Antibody-Mediated Rejection. EXP CLIN TRANSPLANT 2025; 23:192-201. [PMID: 40223384 DOI: 10.6002/ect.2024.0166] [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: 04/15/2025]
Abstract
OBJECTIVES Despite its known role in promoting tolerance, the function of programmed cell death protein 1/programmed death ligand 1 in antibody-mediated rejection is less clear. We aimed to clarify this role by examining expression of programmed cell death protein 1/programmed death ligand 1 in renal allografts diagnosed with antibody-mediated rejection. MATERIALS AND METHODS We examined 110 patients: 68 with pure antibody-mediated rejection (group 1) and 42 with both antibody-mediated rejection and T-cell mediated rejection (group 2). Renal immune cell infiltration, cytokine expression, and programmed cell death protein 1/programmed death ligand 1 expres-sion were examined immunohistochemically. RESULTS Expression of programmed cell death protein 1/programmed death ligand 1 in endothelial and inflammatory cells was higher in group 2 versus in group 1 (P < .001). Expression of programmed cell death protein 1/programmed death ligand 1 increased with immune cell infiltration. An inverse relationship existed between peritubular capillary DR expression and programmed cell death protein 1/programmed death ligand 1 interaction, with a positive correlation with tubular HLA-DR. Development of interstitial fibrosis was shown in 52.3% of patients with endothelial programmed cell death protein 1/programmed death ligand 1 interaction compared with 12.1% without this interaction (P < .001). Ten-year survival rate was 27.3% in patients with versus 66.7% in patients without endothelial programmed cell death protein 1/programmed death ligand 1 (P < .001) and 31.3% in patients with and 66.1% in patients without inflammatory cell programmed cell death protein 1/programmed death ligand 1 expression (P < .001). CONCLUSIONS Heightened immunological nature in antibody-mediated rejection may influence the unexpected functions of programmed death ligand 1. Inhibitory functions of the programmed cell death protein 1/programmed death ligand 1 pathway may be less effective under strong T-cell activation with high immunological costimulation in antibody-mediated rejection.
Collapse
Affiliation(s)
- Binnaz Handan Özdemir
- From the Pathology Department, Başkent University Faculty of Medicine, Ankara, Turkey
| | | | | | | |
Collapse
|
37
|
Jafri Z, Zhang J, O'Meara CH, Joshua AM, Parish CR, Khachigian LM. Interplay between CD28 and PD-1 in T cell immunotherapy. Vascul Pharmacol 2025; 158:107461. [PMID: 39734005 DOI: 10.1016/j.vph.2024.107461] [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: 11/26/2024] [Revised: 12/26/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
Immune checkpoint therapy targeting the PD-1/PD-L1 axis has revolutionised the treatment of solid tumors. However, T cell exhaustion underpins resistance to current anti-PD-1 therapies, resulting in lower response rates in cancer patients. CD28 is a T cell costimulatory receptor that can influence the PD-1 signalling pathway (and vice versa). CD28 signalling has the potential to counter T cell exhaustion by serving as a potential complementary response to traditional anti-PD-1 therapies. Here we discuss the interplay between PD-1 and CD28 in T cell immunotherapy and additionally how CD28 transcriptionally modulates T cell exhaustion. We also consider clinical attempts at targeting CD28; the challenges faced by past attempts and recent promising developments.
Collapse
Affiliation(s)
- Zuhayr Jafri
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jingwen Zhang
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Connor H O'Meara
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia; Division of Head & Neck Oncology and Microvascular Reconstruction, Department of Otolaryngology, Head & Neck Surgery, University of Virginia Health Services, Charlottesville, VA 22903, USA; Department of Otolaryngology, Head & Neck Surgery, Australian National University, Acton, ACT 0200, Australia
| | - Anthony M Joshua
- Kinghorn Cancer Centre, St Vincents Hospital, Sydney and Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Christopher R Parish
- Cancer and Vascular Biology Group, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research, Department of Pathology, School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
38
|
Kim M, Je Y, Chun J, Youn YH, Park H, Nahm JH, Kim J. Helicobacter pylori Eradication Is Associated With a Reduced Risk of Metachronous Gastric Neoplasia by Restoring Immune Function in the Gastric Mucosa. Helicobacter 2025; 30:e70030. [PMID: 40169366 PMCID: PMC11961346 DOI: 10.1111/hel.70030] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Helicobacter pylori infection is a significant contributing factor of gastric cancer. Metachronous neoplasms also pose a risk. The mechanism underlying the impact of H. pylori eradication on preventing metachronous gastric cancer is unclear. This study aimed to investigate immunity changes in gastric mucosa after H. pylori eradication and to identify mechanisms preventing metachronous recurrence. MATERIALS AND METHODS Patients diagnosed with gastric neoplasm and H. pylori infection, who underwent endoscopic resection, were included. Thirty-six cases of metachronous neoplasms occurring after eradication (metachronous group) were compared to 36 controls matched for age, sex, atrophy, and metaplasia (control group). Histological features and immunohistochemical staining for T-cell (CD3, CD4, and CD8) and immune exhaustion (forkhead/winged helix transcription factor and programmed cell death-ligand 1) markers in the non-tumor-bearing mucosa were evaluated. RESULTS In histologic features, glandular atrophy and intestinal metaplasia in the gastric mucosa significantly improved following H. pylori eradication in the control group (p < 0.001, 0.008), whereas they did not improve in the metachronous group (p = 0.449, 0.609). CD8 and CD8/CD3 ratios increased in the control group (p < 0.001, 0.04), but did not show differences in the metachronous group (p = 0.057, 0.245). The CD4/CD3 ratio and programmed cell death-ligand 1/CD4 expression significantly decreased after H. pylori eradication in the control group (p = 0.003, 0.042), but not in the metachronous group (p = 0.54, 0.55). CONCLUSIONS This observational study suggests that H. pylori eradication may prevent the recurrence of gastric neoplasia by improving histological inflammation and overcoming immune exhaustion.
Collapse
Affiliation(s)
- Min‐Jae Kim
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Yeonjin Je
- Graduate School of MedicineYonsei UniversitySeoulKorea
| | - Jaeyoung Chun
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Young Hoon Youn
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Hyojin Park
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Ji Hae Nahm
- Department of Pathology, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| | - Jie‐Hyun Kim
- Department of Internal Medicine, Gangnam Severance HospitalYonsei University College of MedicineSeoulKorea
| |
Collapse
|
39
|
Pham‐Danis C, Chia SB, Scarborough HA, Danis E, Nemkov T, Zaberezhnyy V, Christenson JL, Kleczko EK, Navarro A, Goodspeed A, Bonney EA, Dinarello CA, Marchetti C, Nemenoff RA, Hansen KC, DeGregori J. Inflammation Promotes Aging-Associated Oncogenesis in the Lung. AGING AND CANCER 2025; 6:3-18. [PMID: 40365571 PMCID: PMC12068184 DOI: 10.1002/aac2.12077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/06/2024] [Accepted: 08/13/2024] [Indexed: 05/15/2025]
Abstract
Background Lung cancer is the leading cause of cancer death in the world. While cigarette smoking is the major preventable factor for cancers in general and lung cancer in particular, old age is also a major risk factor. Aging-related chronic, low-level inflammation, termed inflammaging, has been widely documented; however, it remains unclear how inflammaging contributes to increased lung cancer incidence. Aim The aim of this study was to establish connections between aging-associated changes in the lungs and cancer risk. Methods We analyzed public databases of gene expression for normal and cancerous human lungs and used mouse models to understand which changes were dependent on inflammation, as well as to assess the impact on oncogenesis. Results Analyses of GTEx and TCGA databases comparing gene expression profiles from normal lungs, lung adenocarcinoma, and lung squamous cell carcinoma of subjects across age groups revealed upregulated pathways such as inflammatory response, TNFA signaling via NFκB, and interferon-gamma response. Similar pathways were identified comparing the gene expression profiles of young and old mouse lungs. Transgenic expression of alpha 1 antitrypsin (AAT) partially reverses increases in markers of aging-associated inflammation and immune deregulation. Using an orthotopic model of lung cancer using cells derived from EML4-ALK fusion-induced adenomas, we demonstrated an increased tumor outgrowth in lungs of old mice while NLRP3 knockout in old mice decreased tumor volumes, suggesting that inflammation contributes to increased lung cancer development in aging organisms. Conclusions These studies reveal how expression of an anti-inflammatory mediator (AAT) can reduce some but not all aging-associated changes in mRNA and protein expression in the lungs. We further show that aging is associated with increased tumor outgrowth in the lungs, which may relate to an increased inflammatory microenvironment.
Collapse
Affiliation(s)
- Catherine Pham‐Danis
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Shi B. Chia
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Hannah A. Scarborough
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Etienne Danis
- Department of Biomedical InformaticsUniversity of Colorado Anschutz Medical CampusUniversity of Colorado Cancer CenterAuroraColoradoUSA
- University of Colorado Cancer CenterUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Travis Nemkov
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Vadym Zaberezhnyy
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Jessica L. Christenson
- Department of PathologyUniversity of Colorado Anschutz Medical CampusUniversity of Colorado Cancer CenterAuroraColoradoUSA
| | - Emily K. Kleczko
- Department of MedicineUniversity of Colorado Anschutz Medical CampusUniversity of Colorado Cancer CenterAuroraColoradoUSA
| | - Andre Navarro
- Department of MedicineUniversity of Colorado Anschutz Medical CampusUniversity of Colorado Cancer CenterAuroraColoradoUSA
| | - Andrew Goodspeed
- Department of Biomedical InformaticsUniversity of Colorado Anschutz Medical CampusUniversity of Colorado Cancer CenterAuroraColoradoUSA
- University of Colorado Cancer CenterUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Elizabeth A. Bonney
- Department of Obstetrics, Gynecology and Reproductive SciencesLarner College of Medicine, University of VermontBurlingtonVermontUSA
| | - Charles A. Dinarello
- Department of MedicineUniversity of Colorado Anschutz Medical CampusUniversity of Colorado Cancer CenterAuroraColoradoUSA
| | - Carlo Marchetti
- Department of MedicineUniversity of Colorado Anschutz Medical CampusUniversity of Colorado Cancer CenterAuroraColoradoUSA
| | - Raphael A. Nemenoff
- Department of MedicineUniversity of Colorado Anschutz Medical CampusUniversity of Colorado Cancer CenterAuroraColoradoUSA
| | - Kirk C. Hansen
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - James DeGregori
- Department of Biochemistry and Molecular GeneticsUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
- Department of MedicineUniversity of Colorado Anschutz Medical CampusUniversity of Colorado Cancer CenterAuroraColoradoUSA
- Department of Immunology and MicrobiologyUniversity of Colorado Anschutz Medical CampusUniversity of Colorado Cancer CenterAuroraColoradoUSA
| |
Collapse
|
40
|
Bhatt B, Kumar K, Shi H, Ganesan D, Anazodo F, Rathakrishnan A, Zhu H, Wanna A, Jiang C, Jayavelu T, Lokeshwar VB, Pacholczyk R, Munn DH, Sheridan BS, Moskophidis D, Li H, Singh N. UFL1 promotes survival and function of virtual memory CD8 T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae042. [PMID: 40073095 PMCID: PMC11952874 DOI: 10.1093/jimmun/vkae042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 11/30/2024] [Indexed: 03/14/2025]
Abstract
In naïve mice, a fraction of CD8 T cells displaying high affinity for self-MHC peptide complexes develop into virtual memory T (TVM) cells. Due to self-reactivity, TVM cells are exposed to persistent antigenic stimulation, a condition known to induce T cell exhaustion. However, TVM cells do not exhibit characteristics similar to exhausted CD8 T (TEX) cells. Here, we tested the role of the UFL1, E3 ligase of the ufmylation pathway in TVM cells. We show that UFL1 prevents the acquisition of epigenetic, transcriptional, and phenotypic changes in TVM cells that are similar to TEX cells and thus promote their survival and function. UFL1-deficient TVM cells failed to protect mice against Listeria infection. Epigenetic analysis showed higher BATF activity in UFL1-deficient TVM cells. Deletion of BATF and not PD1 decreased inhibitory molecules expression and restored the survival and function of UFL1-deficient TVM cells. Our findings demonstrate a key role of UFL1 in inhibiting the exhaustion of TVM cells and promoting their survival and function.
Collapse
Affiliation(s)
- Brinda Bhatt
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Kunal Kumar
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Huidong Shi
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
- Immunology Center of Georgia, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Dhasarathan Ganesan
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Francis Anazodo
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Aravind Rathakrishnan
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Huabin Zhu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Andrew Wanna
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Chen Jiang
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Tamilselvan Jayavelu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Vinata Bal Lokeshwar
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Rafal Pacholczyk
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - David H Munn
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
- Immunology Center of Georgia, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Brian S Sheridan
- Center for Infectious Diseases, Stony Brook University, Stony Brook, NY, United States
| | - Demetrius Moskophidis
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Honglin Li
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| | - Nagendra Singh
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
- Immunology Center of Georgia, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States
| |
Collapse
|
41
|
Raposo CJ, Yan PK, Chen AY, Majidi S, Hiam-Galvez KJ, Satpathy AT. Functional memory T cells are derived from exhausted clones and expanded by checkpoint blockade. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637523. [PMID: 39990338 PMCID: PMC11844384 DOI: 10.1101/2025.02.10.637523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Immune checkpoint blockade can facilitate tumor clearance by T cells, resulting in long term patient survival. However, the capacity of exhausted CD8+ T cells (Tex), present during chronic antigen exposure, to form memory after antigen clearance remains unclear. Here, we performed longitudinal single cell RNA/T cell receptor sequencing and ATAC-sequencing on antigen-specific T cells after the clearance of chronic lymphocytic choriomeningitis virus (LCMV) infection. These data revealed the formation of a robust population of memory CD8+ T cells that transcriptionally, epigenetically, and functionally resemble central memory T cells (Tcm) that form after clearance of acute infection. To lineage trace the origin and memory recall response of Tex-derived memory clones, we utilized T cell receptor sequencing over the course of primary infection and rechallenge. We show that chronic Tcm are a clonally distinct lineage of Tex derived from progenitor exhausted cells, persist long-term in the absence of antigen, and undergo rapid clonal expansion during rechallenge. Finally, we demonstrate that αPD-L1 immune checkpoint blockade after chronic LCMV infection preferentially expands clones which form Tcm after clearance. Together, these data support the concept that chronically stimulated T cells form bona fide functional memory T cells through an analogous differentiation pathway to acutely stimulated T cells, which may have significant implications for enhancing immune memory to cancer through checkpoint blockade and vaccination.
Collapse
Affiliation(s)
- Colin J. Raposo
- Department of Pathology, Stanford University, Stanford, CA, USA
- Program in Immunology, Stanford University, Stanford, CA, USA
| | - Patrick K. Yan
- Department of Pathology, Stanford University, Stanford, CA, USA
- Program in Immunology, Stanford University, Stanford, CA, USA
| | - Andy Y. Chen
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Saba Majidi
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Ansuman T. Satpathy
- Department of Pathology, Stanford University, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
42
|
Nik Amirah Auni NMA, Mohd Redzwan N, Fauzi AN, Yahya MM, Wong KK. Hypomethylating agents as emerging therapeutics for triple-negative breast cancer. Life Sci 2025; 363:123403. [PMID: 39824347 DOI: 10.1016/j.lfs.2025.123403] [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: 08/31/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Triple-negative breast cancer (TNBC) is recognized as the most aggressive subtype of breast cancer. Epigenetic silencing, such as DNA methylation mediated by DNA methyltransferases (DNMTs) plays key roles in TNBC tumorigenesis. Hypomethylating agents (HMAs) such as azacitidine, decitabine, and guadecitabine are key inhibitors of DNMTs, and accumulating evidence has shown their immunogenicity properties. In this review, the efficacy and anti-tumor immune responses triggered by HMAs in TNBC are presented and discussed. Essentially, overexpression of DNMTs is associated with poor prognosis and reduced TNBC survival rates, and these effects are negated by HMAs. In particular, HMAs could reverse epigenetic silencing of tumor suppressor genes and enhance immune recognition of TNBC cells. Clinical trials of HMAs in TNBCs are limited but early-stage trials indicate that HMAs are safe and tolerable. More clinical studies are required to establish the effectiveness of HMAs against the disease, as supported by preclinical data substantiating their effectiveness especially guadecitabine. Future research should focus on optimizing dosing and exploring combinations with immunotherapies to maximize the potential of HMAs in TNBC treatment.
Collapse
Affiliation(s)
| | - Norhanani Mohd Redzwan
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Agustine Nengsih Fauzi
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Maya Mazuwin Yahya
- Department of Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
43
|
Feng S, Shen Y, Zhang H, Liu W, Feng W, Chen X, Zhang L, Chen J, Lu M, Xue X, Shen X. Human cytomegalovirus tegument protein UL23 promotes gastric cancer immune evasion by facilitating PD-L1 transcription. Mol Med 2025; 31:57. [PMID: 39934685 PMCID: PMC11816993 DOI: 10.1186/s10020-025-01114-8] [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: 09/13/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Immune checkpoint therapy targeting PD-1/PD-L1 has shown promise in treating tumors, however, its clinical benefits are limited to a subset of gastric cancer (GC) patients. Recent research has highlighted a the correlation between PD-L1 expression and the clinical efficacy of anti-PD-1/PD-L1 therapies. Human cytomegalovirus (HCMV) has been implicated in GC, but its specific role in modulating this disease remains elusive. In this study, we analyzed clinical tissue samples using bioinformatics and real-time quantitative polymerase chain reaction (RT-qPCR). We found that GC tissues infected with HCMV presented higher PD-L1 expression compared to those without virus. Furthermore, we demonstrated that HCMV infection enhances PD-L1 expression in GC cells. Cytotoxicity assays revealed that HCMV modulates cancer immune responses via the PD-1/PD-L1 pathway. Mechanistically, we showed that HCMV activates the PI3K-Akt signaling cascade and modulates PD-L1 expression through its tegument protein UL23. Functionally, increased UL23 expression leads to elevated PD-L1 levels, which diminishes tumor cell sensitivity to T-cell-mediated cytotoxicity and triggers T-cell apoptosis. Additionally, in vivo experiments revealed that UL23-induced PD-L1 upregulation inhibits CD8+ T-cell infiltration and reduces the expression of inflammatory factors in tumor microenvironment, ultimately weakening antitumor immunity. Our findings reveal a novel mechanism whereby HCMV and its tegument protein UL23 contribute to cancer immunosuppression through the regulation of PD-L1 expression. This discovery may serve as a potential therapeutic target for enhancing the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Shiyu Feng
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yitian Shen
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Haoke Zhang
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wanfeng Liu
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Weixu Feng
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiuting Chen
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Liang Zhang
- The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiangli Chen
- Traditional Chinese Medical Hospital of Zhuji, Zhuji, Zhejiang, China
| | - Mingdong Lu
- The Second Affiliated Hospital, Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Xue
- Wenzhou Collaborative Innovation Center of Gastrointestinal Cancer in Basic Research and Precision Medicine, Wenzhou Key Laboratory of Cancer-related Pathogens and Immunity, Department of Microbiology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| | - Xian Shen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
44
|
Kastner AL, Marx AF, Dimitrova M, Abreu-Mota T, Ertuna YI, Bonilla WV, Stauffer K, Künzli M, Wagner I, Kreutzfeldt M, Merkler D, Pinschewer DD. Durable lymphocyte subset elimination upon a single dose of AAV-delivered depletion antibody dissects immune control of chronic viral infection. Immunity 2025; 58:481-498.e10. [PMID: 39719711 DOI: 10.1016/j.immuni.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/02/2024] [Accepted: 11/26/2024] [Indexed: 12/26/2024]
Abstract
To interrogate the role of specific immune cells in infection, cancer, and autoimmunity, immunologists commonly use monoclonal depletion antibodies (depletion-mAbs) or genetically engineered mouse models (GEMMs). To generate a tool that combines specific advantages and avoids select drawbacks of the two methods, we engineered adeno-associated viral vectors expressing depletion mAbs (depletion-AAVs). Single-dose depletion-AAV administration durably eliminated lymphocyte subsets in mice and avoided accessory deficiencies of GEMMs, such as marginal zone defects in B cell-deficient animals. Depletion-AAVs can be used in animals of different genetic backgrounds, and multiple depletion-AAVs can readily be combined. Exploiting depletion-AAV technology, we showed that B cells were required for unimpaired CD4+ and CD8+ T cell responses to chronic lymphocytic choriomeningitis virus (LCMV) infection. Upon B cell depletion, CD8+ T cells failed to suppress viremia, and they only helped resolve chronic infection when antibodies dampened viral loads. Our study positions depletion-AAVs as a versatile tool for immunological research.
Collapse
Affiliation(s)
- Anna Lena Kastner
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | | | - Mirela Dimitrova
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Tiago Abreu-Mota
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Yusuf I Ertuna
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Weldy V Bonilla
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Karsten Stauffer
- Department of Biomedicine, University of Basel, 4009 Basel, Switzerland
| | - Marco Künzli
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Ingrid Wagner
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Mario Kreutzfeldt
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1206 Geneva, Switzerland
| | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland; Division of Clinical Pathology, Geneva University Hospital, 1206 Geneva, Switzerland
| | | |
Collapse
|
45
|
Deecke L, Ohlei O, Goldeck D, Homann J, Toepfer S, Demuth I, Bertram L, Pawelec G, Lill CM. Peripheral Immune Profiles in Individuals at Genetic Risk of Amyotrophic Lateral Sclerosis and Alzheimer's Disease. Cells 2025; 14:250. [PMID: 39996723 PMCID: PMC11852917 DOI: 10.3390/cells14040250] [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: 12/20/2024] [Revised: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
The immune system plays a crucial role in the pathogenesis of neurodegenerative diseases. Here, we explored whether blood immune cell profiles are already altered in healthy individuals with a genetic predisposition to amyotrophic lateral sclerosis (ALS) or Alzheimer's disease (AD). Using multicolor flow cytometry, we analyzed 92 immune cell phenotypes in the blood of 448 healthy participants from the Berlin Aging Study II. We calculated polygenic risk scores (PGSs) using genome-wide significant SNPs from recent large genome-wide association studies on ALS and AD. Linear regression analyses were then performed of the immune cell types on the PGSs in both the overall sample and a subgroup of older participants (>60 years). While we did not find any significant associations between immune cell subtypes and ALS and AD PGSs when controlling for the false discovery rate (FDR = 0.05), we observed several nominally significant results (p < 0.05) with consistent effect directions across strata. The strongest association was observed with CD57+ CD8+ early-memory T cells and ALS risk (p = 0.006). Other immune cell subtypes associated with ALS risk included PD-1+ CD8+ and CD57+ CD4+ early-memory T cells, non-classical monocytes, and myeloid dendritic cells. For AD, naïve CD57+ CD8+ T cells and mature NKG2A+ natural killer cells showed nominally significant associations. We did not observe major immune cell changes in individuals at high genetic risk of ALS or AD, suggesting they may arise later in disease progression. Additional studies are required to validate our nominally significant findings.
Collapse
Affiliation(s)
- Laura Deecke
- Institute of Epidemiology and Social Medicine, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany; (L.D.); (J.H.)
| | - Olena Ohlei
- Institute of Epidemiology and Social Medicine, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany; (L.D.); (J.H.)
| | - David Goldeck
- Department of Immunology, University of Tübingen, 72076 Tübingen, Germany (G.P.)
| | - Jan Homann
- Institute of Epidemiology and Social Medicine, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany; (L.D.); (J.H.)
| | - Sarah Toepfer
- Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Ilja Demuth
- Department of Endocrinology and Metabolic Diseases (Including Division of Lipid Metabolism), Charité–Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- BCRT—Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics (LIGA), University of Lübeck, 23562 Lübeck, Germany
| | - Graham Pawelec
- Department of Immunology, University of Tübingen, 72076 Tübingen, Germany (G.P.)
- Health Sciences North Research Institute of Canada, Sudbury, ON P3E 2H3, Canada
| | - Christina M. Lill
- Institute of Epidemiology and Social Medicine, University of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany; (L.D.); (J.H.)
- Ageing and Epidemiology Unit (AGE), School of Public Health, Imperial College London, London W6 8RP, UK
| |
Collapse
|
46
|
Duan W, Zhou Z, Huang Y, Cui Y, Jin X, Liu R, Chen L. Euphorbia helioscopia L. inhibits lung tumorigenesis through alleviating exhausted T cell induced by chronic inflammation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119097. [PMID: 39537116 DOI: 10.1016/j.jep.2024.119097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Euphorbia helioscopia L. (ZQ) is a very effective traditional Chinese medicine for treating pneumonia and lung cancer. However, the effects and mechanisms by which ZQ prevents lung tumorigenesis in the presence of chronic inflammation remain unexplored. AIM To examine the effects and mechanisms of ZQ in alleviating chronic inflammation-induced T cell exhaustion and inhibiting lung tumorigenesis. METHODS A mice model of lung tumorigenesis under chronic inflammation conditions was established by repeated administration of lipopolysaccharide (LPS) and exposure to the tobacco carcinogen nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). Mice were treated with ZQ (0.9, 1.8, and 3.6 g/kg/day) for 25 weeks. Lung pathology and tumor incidence were assessed, and inflammatory cytokine levels in bronchoalveolar lavage fluid (BALF) and serum were measured. Additionally, the proportions of CD3+ T, CD4+ T, and CD8+ T cells and their inhibitory receptors expression were evaluated. In vitro, T cell exhaustion models were induced using inflammatory-conditioned media, followed by treatment with ZQ (0.5, 2, 8 μg/mL). T cell exhaustion markers and characteristics were analyzed, and molecular mechanisms were explored using RNA sequencing and Immunoblotting analysis. RESULTS In vivo, ZQ significantly reduced inflammatory infiltration and lung damage, tumor incidence, number, size, and lung and spleen indices in mice. It also markedly lowered the levels of pro-inflammatory cytokines and immunosuppressive cytokines in BALF and serum. Additionally, ZQ improved the proportions of CD3+ T, CD4+ T, and CD8+ T cells and significantly decreased the expression of inhibitory receptors on CD4+ T and CD8+ T cells in the lung tissues and spleen. In vitro, ZQ effectively alleviated T cell exhaustion induced by the inflammatory environment, marked by reduced expression of inhibitory receptors, increased cytokine secretion, improved proliferation, and enhanced tumoricidal activity. RNA sequencing revealed that ZQ significantly downregulated the JAK-STAT signaling and upregulated stemness-associated pathways. Immunoblotting results indicated that treatment with ZQ markedly reduced the phosphorylation of Signal transducer and activator of transcription 3 (STAT3) and increased the expression of T cell factor -1/7 (TCF1/7). CONCLUSION ZQ inhibits lung tumorigenesis in LPS/NNK-treated mice through alleviating exhausted T cells induced by chronic inflammation, which is attributed to the suppression of STAT3 activation and the maintenance of stemness characteristics in T cells. These findings provide experimental evidence for the potential use of ZQ in preventing and treating lung tumourigenesis in patients with chronic inflammation and the clinical management of lung cancer patients with concomitant chronic inflammation.
Collapse
Affiliation(s)
- Wenbin Duan
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Ziye Zhou
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Yuqing Huang
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Yaru Cui
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| | - Xuhui Jin
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Ronghua Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Lanying Chen
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, 330006, China; Key Laboratory for Evaluation on Anti-Tumor Effect of Chinese Medicine by Strengthening Body Resistance to Eliminate Pathogenic Factors, Nanchang, 330006, China; Key Laboratory of Effective Material Basis of TCM, Jiangxi Province, Jiangxi University of Chinese Medicine, Nanchang, 330006, China.
| |
Collapse
|
47
|
Hilt ZT, Reynaldi A, Steinhilber M, Zhang S, Wesnak SP, Smith NL, Davenport MP, Rudd BD. Recent thymic emigrants are preferentially recruited into the memory pool during persistent infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636722. [PMID: 39975271 PMCID: PMC11839080 DOI: 10.1101/2025.02.06.636722] [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
Cytomegalovirus (CMV) leads to a unique phenomenon known as 'memory inflation,' where antigen-specific memory CD8+ T cells continue to accumulate in the peripheral tissues during the latent stage of infection. However, it is still not clear how the inflating pool of memory CD8+ T cells is generated and maintained. In this study, we used murine cytomegalovirus (MCMV) as a model of persistent infection and fate-mapping mice to determine the dynamics of CD8+ T cell recruitment into the memory pool. We found that neonatal exposure to CMV leads to an expansion of newly made CD8+ T cells (recent thymic emigrants, RTEs), which are maintained in the long-lived memory compartment. In contrast, CD8+ T cells made during the latent phase of infection (mature CD8+ T cells) contribute little to the memory pool. We also observed notable phenotypic differences between RTEs and mature cells. Whereas RTEs present at the time of infection gave rise to more effector memory cells, the cells produced later in infection were biased towards becoming central memory cells. Importantly, the preferential recruitment of RTEs into the effector memory pool also occurs during adult exposure to CMV. Collectively, these data demonstrate that persistent infection expands the RTE population, and timing of infection dictates whether neonatal or adult RTEs are 'locked in' to the memory pool.
Collapse
Affiliation(s)
- Zachary T. Hilt
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Megan Steinhilber
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Shide Zhang
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Samantha P. Wesnak
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Norah L. Smith
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | - Miles P Davenport
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Brian D. Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
48
|
Greene TT, Jo Y, Chiale C, Macal M, Fang Z, Khatri FS, Codrington AL, Kazane KR, Akbulut E, Swaminathan S, Fujita Y, Fitzgerald-Bocarsly P, Cordes T, Metallo C, Scott DA, Zúñiga EI. Metabolic deficiencies underlie reduced plasmacytoid dendritic cell IFN-I production following viral infection. Nat Commun 2025; 16:1460. [PMID: 39920132 PMCID: PMC11805920 DOI: 10.1038/s41467-025-56603-5] [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: 11/11/2024] [Accepted: 01/23/2025] [Indexed: 02/09/2025] Open
Abstract
Type I Interferons (IFN-I) are central to host protection against viral infections, with plasmacytoid dendritic cells (pDC) being the most significant source, yet pDCs lose their IFN-I production capacity following an initial burst of IFN-I, resulting in susceptibility to secondary infections. The underlying mechanisms of these dynamics are not well understood. Here we find that viral infection reduces the capacity of pDCs to engage both oxidative and glycolytic metabolism. Mechanistically, we identify lactate dehydrogenase B (LDHB) as a positive regulator of pDC IFN-I production in mice and humans; meanwhile, LDHB deficiency is associated with suppressed IFN-I production, pDC metabolic capacity, and viral control following infection. In addition, preservation of LDHB expression is sufficient to partially retain the function of otherwise exhausted pDCs, both in vitro and in vivo. Furthermore, restoring LDHB in vivo in pDCs from infected mice increases IFNAR-dependent, infection-associated pathology. Our work thus identifies a mechanism for balancing immunity and pathology during viral infections, while also providing insight into the highly preserved infection-driven pDC inhibition.
Collapse
Affiliation(s)
- Trever T Greene
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Yeara Jo
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Carolina Chiale
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Monica Macal
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Ziyan Fang
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Fawziyah S Khatri
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Alicia L Codrington
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Katelynn R Kazane
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Elizabeth Akbulut
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Shobha Swaminathan
- Department of Medicine, Division of Infectious Disease, The State University of New Jersey, Rutgers, New Jersey Medical School, Newark, NJ, USA
| | - Yu Fujita
- Division of Next-Generation Drug Development, Research Center for Medical Sciences, The Jikei University School of Medicine, Tokyo, Japan
| | | | - Thekla Cordes
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - Christian Metallo
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Sciences, La Jolla, CA, USA
| | - David A Scott
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Elina I Zúñiga
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
49
|
Dai Y, Yu X, Zhao Y, Wei J, Lin D, Wang J, Zhang R, Yuan X, Li S, Huang S, Liu Q, Zhang Z. Targeted Modulation of the Meningeal Lymphatic Reverse Pathway for Immunotherapy of Breast Cancer Brain Metastases. ACS NANO 2025; 19:4830-4844. [PMID: 39818794 DOI: 10.1021/acsnano.4c15860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Treatment of tumor brain metastases remains challenging due to the ineffectiveness of drugs in crossing the blood-brain barrier (BBB). Here, we proposed a potential strategy to target and modulate the meningeal lymphatic system for immunotherapy of breast cancer brain metastases (BCBM) through peripheral administration. CT/fluorescence dual-modality imaging demonstrated that the phospholipid nanoprobe (α-PLNPs) through intracisternal magna injection effectively labeled and long-range tracked the meningeal lymphatic pathway from meningeal lymphatic vessels (MLVs) to periphery drainage cervical lymph nodes (CLNs). Interestingly, the reverse pathway from CLNs to MLVs was also successfully labeled with α-PLNPs through cervical subcutaneous injection, facilitating the noninvasive delivery of immunomodulators to the meningeal lymphatics. Given this, we used melittin-carrying α-M-PLNPs to trigger the modulation of the meningeal lymphatic reverse pathway, which effectively prevents BCBM and prolongs the survival of mice through activating the antigen-presenting cells in the CLNs and promoting the migration of CD8+ T cells into the metastatic brain tumors. This study highlights the potential of the meningeal lymphatic reverse pathway for the immunotherapy of BCBM, which holds great promise for central nervous system disease therapy without the need for drug delivery via BBB.
Collapse
Affiliation(s)
- Yanfeng Dai
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, Hainan 570228, China
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572024, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572024, China
| | - Xiang Yu
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, Hainan 570228, China
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572024, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572024, China
| | - Yifan Zhao
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jianshuang Wei
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Dong Lin
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Jialu Wang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Ren Zhang
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Xuenan Yuan
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Sanmu Li
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572024, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572024, China
| | - Songlin Huang
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, Hainan 570228, China
| | - Qian Liu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572024, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572024, China
| | - Zhihong Zhang
- School of Life and Health Sciences, Hainan Province Key Laboratory of One Health, Collaborative Innovation Center of One Health, Hainan University, Haikou, Hainan 570228, China
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572024, China
- Key Laboratory of Biomedical Engineering of Hainan Province, Collaborative Innovation Center of One Health, Hainan University, Sanya 572024, China
- Britton Chance Center and MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| |
Collapse
|
50
|
Liu Z, Shi Z, Jiang W, Shen Z, Chen W, Shen K, Sun Y, Tang Z, Wang X. Circulating tumor DNA analysis for prediction of prognosis and molecular insights in patients with resectable gastric cancer: results from a prospective study. MedComm (Beijing) 2025; 6:e70065. [PMID: 39830022 PMCID: PMC11742430 DOI: 10.1002/mco2.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 12/03/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
This study aimed to evaluate the prognostic value of plasma circulating tumor DNA (ctDNA) level in patients with resectable gastric cancer (GC). A total of 59 patients were prospectively enrolled, with their ctDNA detected and paired tumor tissue collected at various peri-operative time points. Patients with higher 1-month post-operative ctDNA levels demonstrated shorter overall survival status (hazard ratio [HR] = 5.30, p = 0.0022) and a higher risk of recurrence (HR = 3.85, p = 0.011). The model combining ctDNA with conventional serum tumor markers for GC, including carcinoembryonic antigen, carbohydrate antigen 19-9, and CA72-4, shows high predictive effectiveness for GC prognosis with an area under the curve of 0.940 (p = 0.002), which is higher than net ctDNA and other models without ctDNA. Patients with lower ctDNA levels were more likely to have positive stromal programmed cell death ligand 1 expression (p = 0.046). Additionally, DCAF4L2 mutation was identified as the crucial gene mutation in ctDNA suggesting poor prognosis of patients with GC. Overall, this study highlights that post-operative ctDNA can serve as an effective biomarker for prognostic prediction and recurrence surveillance in resectable GC.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zhongyi Shi
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Wenchao Jiang
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhenbin Shen
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Weidong Chen
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Kuntang Shen
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Yihong Sun
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Department of General SurgeryZhongshan Hospital (Xiamen Branch)Fudan UniversityShanghaiChina
| | - Zhaoqing Tang
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Department of General SurgeryZhongshan Hospital (Xiamen Branch)Fudan UniversityShanghaiChina
| | - Xuefei Wang
- Department of Gastrointestinal SurgeryZhongshan HospitalFudan UniversityShanghaiChina
- Gastric Cancer CenterZhongshan HospitalFudan UniversityShanghaiChina
- Department of General SurgeryZhongshan Hospital (Xiamen Branch)Fudan UniversityShanghaiChina
| |
Collapse
|