1
|
Qiao Y, Xie D, Li Z, Cao S, Zhao D. Global research trends on biomarkers for cancer immunotherapy: Visualization and bibliometric analysis. Hum Vaccin Immunother 2025; 21:2435598. [PMID: 39773010 PMCID: PMC11730411 DOI: 10.1080/21645515.2024.2435598] [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: 09/05/2024] [Revised: 11/08/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
The global burden of cancer continues to grow, posing a significant public health challenge. Although cancer immunotherapy has shown significant efficacy, the response rate is not high. Therefore, the objective of our research was to identify the latest research trends and hotspots on biomarkers from 1993 to 2023. Data were collected from the database Web of Science core collection. Bibliometric analysis and visualization were conducted with CiteSpace(6.3.1), VOSviewer (v1.6.20), R-bibliometrix(v4.3.3), and Microsoft Excel(2019). A total of 2686 literatures were retrieved. The sheer annual volume of publications has shown a rapid upward trend since 2015. The United States has generated the most publications and Harvard University ranked as a leading institution. The global biomarker research on immune checkpoint inhibitors (ICIs) revealed regional differences and in-depth explorations should be promoted in developing countries. Although China has become the second largest country in terms of publication, the average citation per paper and the total link strength were both lower than the other countries. The research on biomarkers mainly concentrated upon the following aspects: PD-1/PD-L1, CTLA-4, gene expression, adverse events, total mutational burden (TMB), body mass index (BMI), gut microbiota, cd8(+)/cd4(+) t-cells, and blood-related biomarkers such as lactate dehydrogenase (LDH), neutrophil-lymphocyte ratio (NLR), cytokines. Furthermore, "artificial intelligence" and "machine learning" have become the most important research hotspot over the last 2 y, which will help us to identify useful biomarkers from complex big data and provide a basis for precise medicine for malignant tumors.
Collapse
Affiliation(s)
- Yuan Qiao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Xie
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhengxiang Li
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Shaohua Cao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Zhao
- Department of Clinical Laboratory, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| |
Collapse
|
2
|
El-Ghazzi N, Monier A, Italiano A, Besson A, Angeli E. Immune-induced thrombocytopenia by pembrolizumab: case report and review of literature. Platelets 2025; 36:2487767. [PMID: 40178025 DOI: 10.1080/09537104.2025.2487767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 02/01/2025] [Accepted: 03/16/2025] [Indexed: 04/05/2025]
Abstract
Immune-checkpoint blockades (ICBs) are now used in early-stage diseases like triple-negative breast cancer (TNBC). While effective, they can cause severe toxicities. We report the first case of life-threatening immune thrombocytopenia (ITP) induced by pembrolizumab during neoadjuvant chemo-immunotherapy for early TNBC. A 42-year-old woman with early-stage TNBC developed grade 4 thrombocytopenia, diagnosed as ITP, after 107 days of pembrolizumab treatment. She required intensive care unit (ICU) admission and high-dose steroids, and intravenous immunoglobulin therapy, leading to a rapid recovery. ITP is a rare but potentially fatal complication of immunotherapy, with an incidence of less than 1% and a mortality rate of up to 20% in affected patients. Immediate recognition and steroid therapy are critical, as platelet transfusion is usually ineffective. Diagnosis is often delayed due to its similarity to chemotherapy-induced marrow toxicity. Immunotherapy-induced ITP generally contraindicates further use of the treatment. ITP, although uncommon, is a serious complication of immunotherapy requiring immediate intervention. The growing use of immunotherapy necessitates increased awareness of its potential toxicities among healthcare providers.
Collapse
Affiliation(s)
- Nathan El-Ghazzi
- Medical Oncology Department, Institut Bergonié, Bordeaux, France
| | - Anna Monier
- Internal Medicine Department, Bordeaux University Hospital, Hôpital Pellegrin, University of Bordeaux, Bordeaux, France
| | - Antoine Italiano
- Medical Oncology Department, Institut Bergonié, Bordeaux, France
| | - Aude Besson
- Medical Oncology Department, Institut Bergonié, Bordeaux, France
| | - Eurydice Angeli
- Medical Oncology Department, Institut Bergonié, Bordeaux, France
| |
Collapse
|
3
|
Huang M, Ji Q, Huang H, Wang X, Wang L. Gut microbiota in hepatocellular carcinoma immunotherapy: immune microenvironment remodeling and gut microbiota modification. Gut Microbes 2025; 17:2486519. [PMID: 40166981 PMCID: PMC11970798 DOI: 10.1080/19490976.2025.2486519] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 03/05/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality, with limited treatment options at advanced stages. The gut microbiota, a diverse community of microorganisms residing in the gastrointestinal tract, plays a pivotal role in regulating immune responses through the gut-liver axis. Emerging evidence underscores its impact on HCC progression and the efficacy of immunotherapy. This review explores the intricate interactions between gut microbiota and the immune system in HCC, with a focus on key immune cells and pathways involved in tumor immunity. Additionally, it highlights strategies for modulating the gut microbiota - such as fecal microbiota transplantation, dietary interventions, and probiotics - as potential approaches to enhancing immunotherapy outcomes. A deeper understanding of these mechanisms could pave the way for novel therapeutic strategies aimed at improving patient prognosis.
Collapse
Affiliation(s)
- Mingyao Huang
- School of Basic Medicine, Putian University, Putian, Fujian, China
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fuzhou, Fujian, China
| | - Quansong Ji
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Huiyan Huang
- Ward 3, De’an Hospital, Xianyou County, Putian, Fujian, China
| | - Xiaoqian Wang
- Department of Rehabilitation Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Lin Wang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
4
|
Tian H, Deng H, Liu X, Liu C, Zhang C, Leong KW, Fan X, Ruan J. A novel FTO-targeting nanodrug induces disulfidptosis and ameliorates the suppressive tumor immune environment to treat uveal melanoma. Biomaterials 2025; 319:123168. [PMID: 40015005 DOI: 10.1016/j.biomaterials.2025.123168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 03/01/2025]
Abstract
Uveal melanoma (UM) is the most prevalent primary ocular malignancy in adults, with high lethality and limited effective treatment options. Despite identified driver mutations in GNAQ, GNA11, and BAP1, therapeutic advancements have been minimal. This study highlights the pivotal role of N6-methyladenosine (m6A) modifications in UM pathogenesis and progression, focusing on the demethylase FTO as a therapeutic target. Elevated FTO expression in UM tissues correlates with decreased m6A levels, increased aggressiveness, and poor prognosis. The FTO inhibitor meclofenamic acid (MA) restored m6A levels, upregulated SLC7A11, and induced disulfidptosis, a unique form of cell death triggered by GSH depletion and NADPH consumption. To address MA's limitations in bioavailability and tumor targeting, we developed an MA-loaded nucleic acid nanodrug (SNAMA). SNAMA demonstrated effective tumor growth inhibition in orthotopic and metastatic UM models through GSH-responsive release and m6A-mediated disulfidptosis activation. Incorporating a PD-L1 aptamer into SNAMA further improved tumor targeting and immune modulation, enhancing therapeutic efficacy. This study identifies FTO as a critical target for UM therapy and introduces SNAMA-apt as a promising nanodrug. The findings offer a foundation for m6A-targeted approaches in UM and other malignancies, addressing bioavailability, targeting, and immune evasion challenges.
Collapse
Affiliation(s)
- Hao Tian
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, PR China; Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Hongpei Deng
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, PR China
| | - Xinlong Liu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Chang Liu
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, PR China
| | - Chuan Zhang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, PR China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, PR China.
| | - Jing Ruan
- Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 20025, PR China; Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
5
|
Li C, Liu J, Ren L, Zhang L, Zhang N, Yan S, Wang Y, Fu S, Wei J, Yue H, Wu Y, Tong M, Shi X, Wang H, Zhao D, Shao Q, Zhang Y, Zhao Y, Hou Y. Design, synthesis, and biological evaluation of 2,4-diaminopyrimidine inhibitors of hematopoietic progenitor kinase 1. Bioorg Med Chem Lett 2025; 123:130242. [PMID: 40246181 DOI: 10.1016/j.bmcl.2025.130242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
Cancer immunotherapy is an emerging anti-cancer strategy that enhances immune circulation by targeting the immune system. Among the various targets, HPK1, a member of the mammalian Ste20-like protein serine/threonine kinase family, serves as a crucial negative regulator of immune-mediated mechanisms, positioning it as a promising target for immunotherapy. Herein, based on the reported HPK1 inhibitors characterized by 2,4-diaminopyrimidine components, four series of derivatives were obtained through structural optimization methods. Compound 10c demonstrates significant inhibitory effects on HPK1 kinase, with an IC50 of 0.09 nM. Additionally, it markedly inhibits the phosphorylation of the downstream adaptor protein SLP76, with an IC50 of 33.74 nM, and effectively stimulates the secretion of the T cell activation marker IL-2, exhibiting an EC50 of 84.24 nM. These findings suggest that compound 10c holds considerable promise for applications in immunotherapy.
Collapse
Affiliation(s)
- Chunting Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Jiuyu Liu
- Department of Biomedical and Chemical Engineering, Liaoning Institute of Science and Technolgy, Benxi 117004, China.
| | - Le Ren
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Long Zhang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Na Zhang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Shaoxuan Yan
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Yu Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Siyu Fu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Jiakuan Wei
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Hao Yue
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Yongshuo Wu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Minghui Tong
- 3D BioOptima, 1338 Wuzhong Avenue, Suzhou 215104, China
| | - Xuan Shi
- 3D BioOptima, 1338 Wuzhong Avenue, Suzhou 215104, China
| | - Han Wang
- 3D BioOptima, 1338 Wuzhong Avenue, Suzhou 215104, China
| | - Dong Zhao
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd., No. 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, China
| | - Qingfeng Shao
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd., No. 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, China
| | - Yuanle Zhang
- Yangtze River Pharmaceutical Group Jiangsu Haici Biological Pharmaceutical Co., Ltd., No. 8 Taizhen Road, Medical New & Hi-tech Industrial Development Zone, Taizhou City, Jiangsu Province, China
| | - Yanfang Zhao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China
| | - Yunlei Hou
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, Liaoning 110016, China..
| |
Collapse
|
6
|
Wang Q, He J, Lei T, Li X, Yue S, Liu C, Hu Q. New insights into cancer immune checkpoints landscape from single-cell RNA sequencing. Biochim Biophys Acta Rev Cancer 2025; 1880:189298. [PMID: 40088992 DOI: 10.1016/j.bbcan.2025.189298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Immune checkpoint blockade (ICB) therapy represents a pivotal advancement in tumor immunotherapy by restoring the cytotoxic lymphocytes' anti-tumor activity through the modulation of immune checkpoint functions. Nevertheless, many patients experience suboptimal therapeutic outcomes, likely due to the immunosuppressive tumor microenvironment, drug resistance, and other factors. Single-cell RNA sequencing has assisted to precisely investigate the immune infiltration patterns before and after ICB treatment, enabling a high-resolution depiction of previously unrecognized functional interaction among immune checkpoints. This review addresses the heterogeneity between tumor microenvironments that respond to or resist ICB therapy, highlighting critical factors underlying the variation in immunotherapy efficacy and elucidating treatment failure. Furthermore, a comprehensive examination is provided of how specific ICBs modulate immune and tumor cells to achieve anti-tumor effects and generate treatment resistance, alongside a summary of emerging immune checkpoints identified as promising targets for cancer immunotherapy through single-cell RNA sequencing applications.
Collapse
Affiliation(s)
- Qian Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiahui He
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tianyu Lei
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaohui Li
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China
| | - Shengqin Yue
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chao Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China.
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Renmin Hospital of Wuhan Economic and Technological Development Zone (Hannan), Wuhan 430090, China.
| |
Collapse
|
7
|
Lei R, Li Q, Wang R, Wu Z, Kong Q, Liu S, Luo Z, Liu X, Zhu X, Wu J. Astragalus injection antagonizes the efficacy of anti-PD-1 against melanoma through down-regulating MHC-II expression. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119966. [PMID: 40354837 DOI: 10.1016/j.jep.2025.119966] [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: 01/03/2025] [Revised: 05/01/2025] [Accepted: 05/10/2025] [Indexed: 05/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragalus injection, a solution derived from Astragalus mongholicus Bunge, has been used in cancer patients for its immune-boosting properties. The effects of Astragalus injection combined with immune checkpoint inhibitors (ICIs) in cancer remain unsuspected. AIM OF THE STUDY This study aims to investigate the efficacy of combining Astragalus injection with anti-PD-1 and to elucidate the potential mechanisms. MATERIALS AND METHODS A melanoma-bearing mouse model was established to observe the effects of Astragalus injection combined with anti-PD-1 treatment. The immune cell infiltration in tumor tissues was evaluated by flow cytometry. Subsequently, network pharmacology and RNA sequencing were conducted to anticipate latent mechanisms of Astragalus in melanoma. Finally, these predictions were validated by flow cytometry, qPCR, Western blotting, and ELISA. RESULTS Astragalus injection significantly antagonized the efficacy of anti-PD-1 in melanoma, resulting in a notable reduction of immune cell infiltration. The network pharmacology and RNA sequencing analysis revealed critical signaling routes, encompassing T-cell receptor activation, immune antigen presentation, and the JAK/STAT pathway. Subsequent data suggested that Astragalus injection could down-regulate the expressions of MHC-II in dendritic cells and B16-OVA cells and restrict the activation of dendritic cells. Moreover, the expressions of CIITA and phosphorylated STAT1 were prominently inhibited by Astragalus injection, whereas the overexpression of STAT1 partially reversed the Astragalus injection-induced decrease of MHC-II. CONCLUSIONS Astragalus antagonized the therapeutic effect of anti-PD-1 in melanoma. These effects were partially through inhibiting JAK/STAT signaling and down-regulating MHC-II expression.
Collapse
Affiliation(s)
- Rui Lei
- Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Qiao Li
- Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Ruilong Wang
- Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Zhuo Wu
- Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Qing Kong
- Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Suqing Liu
- Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Zhuyu Luo
- Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Xiao Liu
- Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Xiaohua Zhu
- Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China.
| | - Jinfeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China; The Second Affiliated Hospital, Yunnan University of Chinese Medicine, Yunan, China.
| |
Collapse
|
8
|
Hou X, Wang C, Zhong Y, Wang L, Kang DD, Lubitz G, Xue Y, Liu Z, Wang S, Li H, Tian M, Cao D, Guo K, Deng B, McComb DW, Marron TU, Brown BD, Merad M, Brody JD, Dong Y. Enhancing antitumor immunity through chemotherapeutic-derived lipid nanoparticle-induced immunogenic cell death and CD40L/Flt3L mRNA-mediated dendritic cell activation. J Control Release 2025; 382:113684. [PMID: 40185331 DOI: 10.1016/j.jconrel.2025.113684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Dendritic cells (DCs) are essential for inducing effective antitumor T cell responses. However, the immunosuppressive tumor microenvironment (TME) hinders DC recruitment and maturation, facilitating tumor progression and spread. This study investigates the synergistic potential of immunogenic cell death (ICD), triggered by chemotherapeutic-derived lipid nanoparticles (LNPs), in combination with Flt3L and CD40L mRNA delivery to enhance DC mobilization and activation, reprogram the TME, and ultimately promote robust antitumor T cell responses. The optimized LNP formulation, GEM5Q7, efficiently delivered mRNA and induced ICD in melanoma cells. Intratumoral administration of GEM5Q7, encapsulating Flt3L and CD40L mRNAs, elevated pro-inflammatory cytokine and chemokine secretion, driving the infiltration and activation of cross-presenting DCs, which are critical for priming T cells. In a subcutaneous melanoma model, this approach led to significant tumor suppression and a 40 % complete response rate. This strategy holds promise for enhancing cancer immunotherapies by reprogramming the TME and inducing durable antitumor T cell immunity.
Collapse
Affiliation(s)
- Xucheng Hou
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chang Wang
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yichen Zhong
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Leiming Wang
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Diana D Kang
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Gabrielle Lubitz
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yonger Xue
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhengwei Liu
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Siyu Wang
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Haoyuan Li
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Meng Tian
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dinglingge Cao
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kaiyuan Guo
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Binbin Deng
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH 43212, USA
| | - David W McComb
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, OH 43212, USA
| | - Thomas U Marron
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian D Brown
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joshua D Brody
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Yizhou Dong
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York 10029, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
9
|
Chen X, Yuan Y, Zhou F, Li L, Pu J, Zeng Y, Jiang X. Lactylation: From Homeostasis to Pathological Implications and Therapeutic Strategies. MedComm (Beijing) 2025; 6:e70226. [PMID: 40443721 PMCID: PMC12122191 DOI: 10.1002/mco2.70226] [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: 09/25/2024] [Revised: 04/16/2025] [Accepted: 04/24/2025] [Indexed: 06/02/2025] Open
Abstract
Lactylation, a recently identified post-translational modification, represents a groundbreaking addition to the epigenetic landscape, revealing its pivotal role in gene regulation and metabolic adaptation. Unlike traditional modifications, lactylation directly links metabolic intermediates, such as lactate, to protein function and cellular behavior. Emerging evidence highlights the critical involvement of lactylation in diverse biological processes, including immune response modulation, cellular differentiation, and tumor progression. However, its regulatory mechanisms, biological implications, and disease associations remain poorly understood. This review systematically explores the enzymatic and nonenzymatic mechanisms underlying protein lactylation, shedding light on the interplay between cellular metabolism and epigenetic control. We comprehensively analyze its biological functions in normal physiology, such as immune homeostasis and tissue repair, and its dysregulation in pathological contexts, including cancer, inflammation, and metabolic disorders. Moreover, we discuss advanced detection technologies and potential therapeutic interventions targeting lactylation pathways. By integrating these insights, this review aims to bridge critical knowledge gaps and propose future directions for research. Highlighting lactylation's multifaceted roles in health and disease, this review provides a timely resource for understanding its clinical implications, particularly as a novel target for precision medicine in metabolic and oncological therapies.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan ProvinceThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingChina
| | - Yixiao Yuan
- Department of Medicine, UF Health Cancer CenterUniversity of FloridaGainesvilleFloridaUSA
| | - Fan Zhou
- Department of Hematologythe Second Hospital Affiliated to Kunming Medical UniversityKunmingChina
| | - Lihua Li
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingChina
| | - Jun Pu
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan ProvinceThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
- NHC Key Laboratory of Drug Addiction MedicineKunming Medical UniversityKunmingChina
| | - Yong Zeng
- Key Laboratory of Neurological and Psychiatric Disease Research of Yunnan ProvinceThe Second Affiliated Hospital of Kunming Medical UniversityKunmingChina
| | - Xiulin Jiang
- Department of Medicine, UF Health Cancer CenterUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
10
|
Jayson A, Tsambarlis M, Thakkar H, Ludlow SP, Syed M. Multisystem immune-related adverse reactions with immune checkpoint inhibitors. JAAPA 2025; 38:29-31. [PMID: 40421943 DOI: 10.1097/01.jaa.0000000000000211] [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: 05/28/2025]
Abstract
ABSTRACT Immune checkpoint inhibitors have transformed oncologic practice, producing durable responses in many patients with advanced malignancies. However, these treatments can cause immune-related adverse reactions. This article describes a patient with metastatic non-small cell lung cancer who developed toxicities involving multiple organ systems shortly after receiving combined ipilimumab and nivolumab. Although toxicity is more common in just one organ system, multisystem adverse reactions are possible, and clinicians who recognize and manage these reactions promptly can improve patient outcomes.
Collapse
Affiliation(s)
- Amy Jayson
- At H. Lee Moffitt Cancer Center and Research Institute in Tampa, Fla., Amy Jayson, Maria Tsambarlis, and Hardik Thakkar practice in internal and hospital medicine; Steven P. Ludlow practices in pharmacy; and Misbahuddin Syed practices in internal and hospital medicine. The authors have disclosed no potential conflicts of interest, financial or otherwise
| | | | | | | | | |
Collapse
|
11
|
Zhang Y, Chen Y, Guo C, Li S, Huang C. Systemic immune-inflammation index as a predictor of survival in non-small cell lung cancer patients undergoing immune checkpoint inhibition: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2025; 210:104669. [PMID: 39978427 DOI: 10.1016/j.critrevonc.2025.104669] [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: 10/31/2024] [Revised: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND This meta-analysis aims to evaluate the association between pretreatment systemic immune-inflammation index (SII) levels and progression-free survival (PFS) and overall survival (OS) in NSCLC patients receiving immune checkpoint inhibitors (ICIs). METHODS A systematic search was conducted across PubMed, Embase, and Web of Science. Hazard ratios (HRs) with 95 % confidence intervals (CIs) for PFS and OS were extracted or calculated. Random-effects models were employed to pool the results and subgroup analyses were performed based on study characteristics, treatment regimens, and analytical methods. RESULTS Two prospective and 11 retrospective studies involving 2342 NSCLC patients treated with ICIs were included. A high pretreatment SII was significantly associated with poor PFS (HR: 2.05, 95 % CI: 1.59-2.64, p < 0.001; I2 = 42 %) and poor OS (HR: 1.54, 95 % CI: 1.29-1.82, p < 0.001; I2 = 22 %). Subgroup analyses according to the country of the study, lines of treatment, cancer stage, methods for determining the cutoffs of SII, and the analytic models showed consistent results (p for subgroup difference all > 0.05). Interestingly, the subgroup analyses indicated a stronger association in patients receiving ICIs alone versus those receiving concurrent chemotherapy (p for subgroup difference = 0.04). CONCLUSIONS High pretreatment SII is associated with worse PFS and OS in NSCLC patients treated with ICIs, particularly for the patients receiving ICIs alone without concurrent chemotherapy.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yeye Chen
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Chao Guo
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| | - Cheng Huang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
12
|
Liu D, Liu L, Zhao X, Zhang X, Chen X, Che X, Wu G. A comprehensive review on targeting diverse immune cells for anticancer therapy: Beyond immune checkpoint inhibitors. Crit Rev Oncol Hematol 2025; 210:104702. [PMID: 40122356 DOI: 10.1016/j.critrevonc.2025.104702] [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: 02/10/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, primary resistance and acquired resistance continue to limit their efficacy for many patients. To address resistance and enhance the anti-tumor activity within the tumor immune microenvironment (TIME), numerous therapeutic strategies targeting both innate and adaptive immune cells have emerged. These include combination therapies with ICIs, chimeric antigen receptor T-cell (CAR-T), chimeric antigen receptor macrophages (CAR-Ms) or chimeric antigen receptor natural killer cell (CAR-NK) therapy, colony stimulating factor 1 receptor (CSF1R) inhibitors, dendritic cell (DC) vaccines, toll-like receptor (TLR) agonists, cytokine therapies, and chemokine inhibition. These approaches underscore the significant potential of the TIME in cancer treatment. This article provides a comprehensive and up-to-date review of the mechanisms of action of various innate and adaptive immune cells within the TIME, as well as the therapeutic strategies targeting each immune cell type, aiming to deepen the understanding of their therapeutic potential.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinming Zhao
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoman Zhang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaochi Chen
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
13
|
Wu Y, Tao Q, Xie J, Liu X, Zhou Y, Wei C, Zhang C, Wang J, Jin Y. Indole-3-carbinol inhibits PD-L1-mediated immune evasion in hepatocellular carcinoma via suppressing NF-κB p105 Ubiquitination. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156692. [PMID: 40215823 DOI: 10.1016/j.phymed.2025.156692] [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: 12/05/2024] [Revised: 03/10/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide, and immunotherapy has demonstrated significant therapeutic benefit in HCC. Indole-3-carbinol (I3C), a naturally occurring ingredient of cruciferous vegetables, significantly inhibits the growth of a wide range of tumors. However, its mechanism of action has not been fully elucidated. PURPOSE This study aims to verify and explore the immunomodulatory effect of I3C in HCC models, and to investigate the specific role and mechanism by which I3C affects PD-L1 expression through the ubiquitination of NF-κB p105. METHODS In vitro, I3C was treated with HepG2 cells and relevant indicators were analyzed. In vivo, the mouse HCC model was established and the effect of I3C on tumors and immune function was evaluated. Subsequently, the downstream target of I3C was found through target prediction, molecular docking, and molecular dynamics simulation. Finally, combined therapy was used to further investigate the effect of I3C on mouse HCC tumors. RESULTS We observed that I3C resulted in decreased programmed cell death ligand 1 (PD-L1) expression in HepG2 cells and increased CD8 T cell infiltration in tissues. Subsequently, target prediction and molecular docking demonstrated that I3C was able to efficiently bind to NF-κB p105. In addition, overexpression of NF-κB p105 upregulated PD-L1 expression and almost completely eliminated the inhibitory effect of I3C. Notably, the combination of I3C and PD-L1 monoclonal antibodies showed synergistic anti-tumor effects in the mouse HCC model. CONCLUSION This study demonstrated that I3C inhibits PD-L1-mediated immune evasion in HCC via suppressing NF-κB p105 ubiquitination. The role of I3C in tumors deserves further investigation and provides the foundation for the future development of novel immunotherapeutic drugs.
Collapse
Affiliation(s)
- Yongkang Wu
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Qing Tao
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Jing Xie
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Xiao Liu
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Yuanzhi Zhou
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Chengyan Wei
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Chunwei Zhang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Jingjing Wang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China
| | - Yong Jin
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, 230032, Hefei, PR China; Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of National Education, Anhui Medical University, 230032, Hefei, PR China.
| |
Collapse
|
14
|
Ding Y, Ye Z, Ding B, Feng S, Zhang Y, Shen Y. Identification of CXCL13 as a Promising Biomarker for Immune Checkpoint Blockade Therapy and PARP Inhibitor Therapy in Ovarian Cancer. Mol Biotechnol 2025; 67:2428-2442. [PMID: 38856873 DOI: 10.1007/s12033-024-01207-5] [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: 02/13/2024] [Accepted: 05/27/2024] [Indexed: 06/11/2024]
Abstract
Ovarian cancer has poor response rates to immune checkpoint blockade (ICB) therapy, despite the use of genomic sequencing to identify molecular targets. Homologous recombination deficiency (HRD) is a conventional indicator of genomic instability (GI) and has been used as a marker for targeted therapies. Indicators reflecting HRD status have shown potential in predicting the efficacy of ICB treatment. Public databases, including TCGA, ICGC, and GEO, were used to obtain data. HRD scores, neoantigen load, and TMB were obtained from the TCGA cohort. Candidate biomarkers were validated in multiple databases, such as the Imvigor210 immunotherapy cohort and the open-source single-cell sequencing database. Immunohistochemistry was performed to further validate the results in independent cohorts. CXCL10, CXCL11, and CXCL13 were found to be significantly upregulated in HRD tumors and exhibited prognostic value. A comprehensive analysis of the tumor immune microenvironment (TIME) revealed that CXCL13 expression positively correlated with neoantigen load and immune cell infiltration. In addition, single-cell sequencing data and clinical trial results supported the utility of CXCL13 as a biomarker for ICB therapy. Not only does CXCL13 serve as a biomarker reflecting HRD status, but it also introduces a potentially novel perspective on prognostic biomarkers for ICB in ovarian cancer.
Collapse
Affiliation(s)
- Yue Ding
- Department of Obstetrics and Gynaecology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Zheng Ye
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Bo Ding
- Department of Obstetrics and Gynaecology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Songwei Feng
- Department of Obstetrics and Gynaecology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Yang Zhang
- Department of Obstetrics and Gynecology, First People's Hospital of Lianyungang, No. 6 East Zhenhua Road, Haizhou, Lianyungang, China.
| | - Yang Shen
- Department of Obstetrics and Gynaecology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
15
|
Güven DC, Thong MS, Arndt V. Survivorship outcomes in patients treated with immune checkpoint inhibitors: a scoping review. J Cancer Surviv 2025; 19:806-845. [PMID: 38175366 PMCID: PMC12081552 DOI: 10.1007/s11764-023-01507-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have become a central part of cancer care. However, the survivorship outcomes in patients treated with ICIs are understudied. Therefore, we conducted a scoping review to evaluate the current status of the field and to establish research gaps regarding survivorship outcomes with ICIs in real-life cohorts. METHODS We used the Web of Science, PubMed, and Embase databases to systematically filter published studies with real-life cohorts from January 1, 2010, until October 19, 2022. Studies evaluating at least one survivorship outcome in ICI-treated patients were included. RESULTS A total of 39 papers were included. Quality of life (QoL) (n = 23), toxicity burden (n = 16), and psychosocial issues (n = 9) were the most frequently evaluated survivorship outcomes. Anti-PD-1/PD-L1 monotherapy and a response to treatment were associated with better QoL. In addition, the ICIs were associated with grade 3 or higher immune-related adverse events (irAEs) in 10-15% and late/long-term irAEs in 20-30% of the survivors. Regarding psychosocial problems, over 30% of survivors showed evidence of anxiety and depression, and 30-40% of survivors reported neurocognitive impairments. CONCLUSION The survivors treated with ICIs have impairments in most survivorship domains. Further research is needed to gather data on the understudied survivorship outcomes like late and long-term effects, fertility, financial toxicity, and return to work in survivors treated with ICIs. IMPLICATIONS FOR CANCER SURVIVORS Available evidence demonstrates that a significant portion of survivors treated with ICIs have a significant toxicity burden, lower QoL than the general population, and a high rate of psychosocial problems.
Collapse
Affiliation(s)
- Deniz Can Güven
- Department of Medical Oncology, Hacettepe University Cancer Institute, 06100 Sihhiye, Ankara, Turkey.
- Health Sciences University, Elazig City Hospital, Elazig, Turkey.
- Unit of Cancer Survivorship, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Melissa Sy Thong
- Unit of Cancer Survivorship, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Volker Arndt
- Unit of Cancer Survivorship, Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
16
|
Jain M, Jadhav IM, Dangat SV, Singuru SR, Sethi G, Yuba E, Gupta RK. Overcoming the novel glycan-lectin checkpoints in tumor microenvironments for the success of the cross-presentation-based immunotherapy. Biomater Sci 2025. [PMID: 40421610 DOI: 10.1039/d4bm01732c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
In pursuit of meeting the ever-rising demand for cancer therapies, cross-presentation-based glyconanovaccines (GNVs) targeting C-type lectin receptors (CLRs) on DCs have shown significant potential as cutting-edge cancer immunotherapy. GNVs are an attractive approach to induce anti-cancer cytotoxic T lymphocyte responses. Despite immune checkpoints (ICs) being well established and an obstacle to the success of GNVs, glycan-lectin circuits are emerging as unique checkpoints due to their immunomodulatory functions. Given the role of aberrant tumor glycosylation in promoting immune evasion, mitigating these effects is crucial for the efficacy of GNVs. Lectins, such as siglecs and galectins, are detrimental to the tumor immune landscape as they promote an immunosuppressive TME. From this perspective, this review aims to explore glycan-lectin ICs and their influence on the efficacy of GNVs. We aim to discuss various ICs in the TME followed by drawbacks of immune checkpoint inhibitors (ICIs). We will also emphasize the altered glycosylation profile of tumors, addressing their immunosuppressive nature along with ways in which CLRs, siglecs, and galectins contribute to immune evasion and cancer progression. Considering the resistance towards ICIs, current and prospective approaches for targeting glycan-lectin circuits and future prospects of these endeavors in harnessing the full potential of GNVs will also be highlighted.
Collapse
Affiliation(s)
- Mannat Jain
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Isha M Jadhav
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Suyash Vinayak Dangat
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Srinivasa Rao Singuru
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| | - Eiji Yuba
- Department of Chemistry & Bioengineering, Graduate School of Engineering, Osaka Metropolitan University, 3-3-138 Sugimoto, Sumiyoshi-ku, Osaka-city, Osaka 558-8585, Japan.
| | - Rajesh Kumar Gupta
- Protein Biochemistry Research Centre, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Tathawade, Pune-411033, Maharashtra, India.
| |
Collapse
|
17
|
Luo T, Guo J, Xi J, Luo X, Fu Z, Chen W, Huang D, Chen K, Xiao Q, Wei S, Wang Y, Du H, Liu L, Cai S, Dong H. Development and validation of a CT-based radiomics machine learning model for differentiating immune-related interstitial pneumonia. Int Immunopharmacol 2025; 156:114681. [PMID: 40262251 DOI: 10.1016/j.intimp.2025.114681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/20/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
INTRODUCTION Immune checkpoint inhibitor-related interstitial pneumonia (CIP) poses a diagnostic challenge due to its radiographic similarity to other pneumonias. We developed a non-invasive model using CT imaging to differentiate CIP from other pneumonias (OTP). METHODS We analyzed CIP and OTP patients after the immunotherapy from five medical centers between 2020 and 2023, and randomly divided into training and validation in 7:3. A radiomics model was developed using random forest analysis. A new model was then built by combining independent risk factors for CIP. The models were evaluated using ROC, calibration, and decision curve analysis. RESULTS A total of 238 patients with pneumonia following immunotherapy were included, with 116 CIP and 122 OTP. After random allocation, the training cohort included 166 patients, and the validation included 72 patients. A radiomics model composed of 11 radiomic features was established using the random forest method, with an AUC of 0.833 for the training cohort and 0.821 for the validation. Univariate and multivariate logistic regression analysis revealed significant differences in smoking history, radiotherapy history, and radiomics score between CIP and OTP (p < 0.05). A new model was constructed based on these three factors and a nomogram was drawn. This model showed good calibration and net benefit in both the training and validation cohorts, with AUCs of 0.872 and 0.860, respectively. CONCLUSION Using the random forest method of machine learning, we successfully constructed a CT-based radiomics CIP differential diagnostic model that can accurately, non-invasively, and rapidly provide clinicians with etiological support for pneumonia diagnosis.
Collapse
Affiliation(s)
- Tingyue Luo
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jingze Guo
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junjie Xi
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoyu Luo
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Zeyu Fu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weisheng Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Danhui Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kaijun Chen
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiang Xiao
- Pulmonary and Critical Care Medicine, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Shuquan Wei
- Department of Pulmonary and Critical Care Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangdong, China
| | - Yan Wang
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Huijuan Du
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Laiyu Liu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China..
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China..
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China..
| |
Collapse
|
18
|
Chen X, Yang M, Zhang H, Wang Y, Yan W, Cheng C, Guo R, Chai J, Zheng Y, Zhang F. Cucurbitacin B induces oral squamous cell carcinomapyroptosis via GSDME and inhibits tumour growth. Transl Oncol 2025; 58:102422. [PMID: 40424936 DOI: 10.1016/j.tranon.2025.102422] [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: 10/18/2024] [Revised: 05/12/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Pyroptosis, a form of programmed cell death, has been shown to induce anti-tumour immunity and inhibit tumour growth. Oral squamous cell carcinoma (OSCC), a prevalent malignant tumour, could benefit from pyroptosis induction as a therapeutic strategy. Cucurbitacin B (CuB), a natural compound derived from various plants, exhibits broad anti-tumour activity. However, whether CuB can exert its anti-tumour effects in OSCC through pyroptosis remains unexplored. RESULTS CuB significantly inhibited the proliferation of OSCC cells, induced pyroptosis, and elevated the levels of inflammatory factors in the cell supernatant. Bioinformatics analysis predicted the potential role of pyroptosis in OSCC, which was subsequently validated in a 4NQO-induced OSCC mouse model. The results demonstrated that CuB not only exerted tumour-inhibitory effects but also increased the infiltration of CD8+ T cells in the peritumoural region. To elucidate the mechanism of CuB-induced pyroptosis, STAT3 was identified as a key target of CuB in OSCC, with its expression upregulated in tumour tissues. Further experiments revealed that CuB induced pyroptosis by suppressing STAT3 expression and promoting the cleavage of caspase-3 and Gasdermin-E (GSDME). CONCLUSION CuB triggers OSCC pyroptosis through the STAT3/caspase-3/GSDME pathway, enhancing peritumoural CD8+ T cell infiltration and offering a novel strategy to boost tumour immunotherapy efficacy.
Collapse
Affiliation(s)
- Xin Chen
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Mengyuan Yang
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Heng Zhang
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Yajun Wang
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Wenpeng Yan
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Chen Cheng
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Rongrong Guo
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - Jiawei Chai
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China
| | - YaHsin Zheng
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Fang Zhang
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, No.63 Xinjian South Road, Yingze District, Taiyuan, Shanxi 030001, China.
| |
Collapse
|
19
|
Ghorbani Vanan A, Nami MT, Ghorbaninezhad F, Eini P, Bagheri K, Mohammadlou M, Mohammadi F, Tahmasebi S, Safarzadeh E. Macrophage polarization in hepatocellular carcinoma: a lncRNA-centric perspective on tumor progression and metastasis. Clin Exp Med 2025; 25:173. [PMID: 40413657 DOI: 10.1007/s10238-025-01711-1] [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: 03/28/2025] [Accepted: 05/01/2025] [Indexed: 05/27/2025]
Abstract
Hepatocellular carcinoma (HCC) represents a multifaceted and aggressive cancer frequently associated with chronic inflammation and immune cell activation. The pathogenesis of HCC is influenced by a variety of factors such as long non-coding RNAs (lncRNAs). LncRNAs, a significant class of non-coding RNAs, contribute to the intricate nature of the transcriptome and are extensively distributed across various tissues and cell types in mammals. In HCC, these transcripts are crucial not only for deepening our molecular understanding but also for advancing clinical outcomes, as they serve as both oncogenes and tumor suppressors by dysregulating essential genes and signaling pathways. Additionally, macrophage polarization is crucial in HCC tumor progression. The study explores the role of lncRNAs in hepatocellular carcinoma (HCC) and elucidates the specific molecular mechanisms by which key lncRNAs such as HULC and MALAT1 regulate macrophage polarization in the tumor microenvironment. These lncRNAs modulate cytokine profiles and influence immune regulators including IL-10 and TGF-β, steering macrophages toward an M2-like, pro-tumor phenotype that fosters aggressive tumor characteristics and progression. Mechanistically, these transcripts interact with epigenetic modifiers like EZH2 to alter histone modifications and chromatin accessibility, while also stabilizing mRNAs that encode inflammatory mediators, thereby reinforcing an immunosuppressive response. The clinical implications of these findings are substantial. The detection of such lncRNAs in patient samples offers a minimally invasive diagnostic avenue, while their pivotal role in complex immune cell behavior positions them as promising prognostic biomarkers. Moreover, targeting these lncRNAs may lead to innovative therapeutic strategies aimed at disrupting tumor-supportive inflammatory cascades and restoring an effective antitumor immune response. Understanding the intricate interplay between lncRNA-mediated epigenetic regulation and macrophage polarization not only refines our grasp of HCC progression but also opens new pathways for interventions designed to improve patient outcomes.
Collapse
Affiliation(s)
- Ahmad Ghorbani Vanan
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Taha Nami
- Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Farid Ghorbaninezhad
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Pooya Eini
- Toxicological Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamyar Bagheri
- Student Research Committee, Abadan University of Medical Sciences, Abadan, Iran
| | - Maryam Mohammadlou
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
20
|
Harel M, Dahan N, Lahav C, Jacob E, Elon Y, Puzanov I, Kelly RJ, Shaked Y, Leibowitz R, Carbone DP, Gandara DR, Dicker AP. Decoding resistance to immune checkpoint inhibitors in non-small cell lung cancer: a comprehensive analysis of plasma proteomics and therapeutic implications. J Immunother Cancer 2025; 13:e011427. [PMID: 40404205 PMCID: PMC12097049 DOI: 10.1136/jitc-2024-011427] [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: 01/13/2025] [Accepted: 05/05/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have shown substantial benefit for patients with advanced non-small cell lung cancer (NSCLC). However, resistance to ICIs remains a major clinical challenge. Here, we perform a comprehensive bioinformatic analysis of plasma proteomic profiles to explore the underlying biology of treatment resistance in NSCLC. METHODS The analysis was performed on 388 "resistance-associated proteins" (RAPs) that were previously described as pretreatment plasma proteomic predictors within the PROphet computational model designed to predict ICI clinical benefit in NSCLC. Putative tissue origins of the RAPs were explored using publicly available datasets. Enrichment analyses were performed to investigate RAP-related biological processes. Plasma proteomic data from 50 healthy subjects and 272 patients with NSCLC were compared, where patients were classified as displaying clinical benefit (CB; n=76) or no CB (NCB; n=196). Therapeutic agents targeting RAPs were identified in drug and clinical trial databases. RESULTS The RAP set was significantly enriched with proteins associated with lung cancer, liver tissue, cell proliferation, extracellular matrix, invasion, and metastasis. Comparison of RAP expression in healthy subjects and patients with NSCLC revealed five distinct RAP subsets that provide mechanistic insights. The RAP subset displaying a pattern of high expression in the healthy population relative to the NSCLC population included multiple proteins associated with antitumor activities, while the subset displaying a pattern of highest expression in the NCB population included proteins associated with various hallmarks of treatment resistance. Analysis of patient-specific RAP profiles revealed inter-patient diversity of potential resistance mechanisms, suggesting that RAPs may aid in developing personalized therapeutic strategies. Furthermore, examination of drug and clinical trial databases revealed that 17.5% of the RAPs are drug targets, highlighting the RAP set as a valuable resource for drug development. CONCLUSIONS The study provides insight into the underlying biology of ICI resistance in NSCLC and highlights the potential clinical value of RAP profiles for developing personalized therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- The Roswell Park Comprehensive Cancer Center Data Bank and BioRepository, Buffalo, New York, USA
| | - Ronan J Kelly
- Department of Hematology and Oncology, Baylor University Medical Center at Dallas, Dallas, Texas, USA
| | - Yuval Shaked
- Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | | | | | - David R Gandara
- Division of Hematology/Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California, USA
| | - Adam P Dicker
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Keshavarz Sadegh R, Saleki K, Rezaei N. Immune checkpoint inhibitor (ICI) therapy in central nervous system cancers: State-of-the-art and future outlook. Int Immunopharmacol 2025; 159:114837. [PMID: 40394797 DOI: 10.1016/j.intimp.2025.114837] [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: 01/11/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Invasive central nervous system (CNS) cancers are an area where the development of breakthrough therapies is urgently needed. For instance, conditions such as glioblastoma multiforme (GBM) are associated with poor clinical prognosis, with the majority of trials offering no improvement to marginally enhanced survival. Unleashing the potential of targeting the immune system in CNS cancers has gained attention in recent years. Inhibition of immune checkpoints such as CTLA-4, PD-1/PD-L1, TIM-3, and LAG-3 has been attempted in recent trials. While potentially offering a notable edge over other immunotherapies, multi-organ adverse events have been found with the administration of immune checkpoint inhibitors (ICIs). The present review captures the state-of-the-art evidence on ICI treatments in different CNS cancers. Also, we discuss the value of combinational therapies involving ICIs as well as next-generation therapeutics such as bispecific antibodies targeting PD-1/LAG-3/TIM-3 and CRISPR-Cas9-edited PD-1-knock-out checkpoint-resistant CAR T-cells.
Collapse
Affiliation(s)
- Roghaye Keshavarz Sadegh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; USERN MUBabol Office, Universal Scientific Education and Research Network (USERN), Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Xu L, Xiao T, Chao T, Xiong H, Yao W. From genes to therapy: a lipid Metabolism-Related genetic risk model predicts HCC outcomes and enhances immunotherapy. BMC Cancer 2025; 25:895. [PMID: 40389832 PMCID: PMC12090435 DOI: 10.1186/s12885-025-14306-6] [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: 10/25/2024] [Accepted: 05/09/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Hepatocellular Carcinoma (HCC) is related to dysregulated lipid metabolism and immunosuppressive microenvironment. This study developed a genetic risk model using lipid metabolism-related genes to predict survival and immune patterns in HCC patients. METHODS Differentially expressed genes (DEGs) related to lipid metabolism were identified in HCC via the TCGA-LIHC dataset. A risk model for survival prediction was constructed via DEGs related to survival. The immune signature associated with the risk model was also evaluated by the CIBERSORT algorithm, tumor immune dysfunction and exclusion algorithm, and single sample gene set enrichment analysis. RESULTS This study identified six lipid metabolism-related genes, ADH4, LCAT, CYP2C9, CYP17A1, LPCAT1, and ACACA, to construct a lipid metabolism-related gene risk model that can divide HCC patients into low- and high-risk groups. Internal and external validation verified that the risk model could be a signature that could effectively predict HCC patient prognosis. High-risk patients showed disrupted immune cell profiles, reduced tumor-killing capacity, and increased expression of immune checkpoint genes. However, they responded more favorably to immune checkpoint inhibitor (ICB) therapy. The top ten hub genes related to the risk model were associated with tumor progression and deteriorating prognosis. In vitro experiments verified that the downregulation of the top 1 hub gene CDK1 was correlated to the HCC cell proliferation. CONCLUSION The risk model constructed using lipid metabolism-related genes could effectively predict prognosis and was related to the immunosuppressive microenvironment and ICB immunotherapy. The hub genes related to the risk model were potential therapeutic targets.
Collapse
Affiliation(s)
- Lei Xu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ting Xiao
- Department of Ultrasonography, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Wei Yao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
23
|
Yang Y, Zhu L, Xu Y, Liang L, Liu L, Chen X, Li H, Liu H. The progress and prospects of targeting the adenosine pathway in cancer immunotherapy. Biomark Res 2025; 13:75. [PMID: 40390144 PMCID: PMC12090549 DOI: 10.1186/s40364-025-00784-0] [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: 02/08/2025] [Accepted: 04/26/2025] [Indexed: 05/21/2025] Open
Abstract
Despite the notable success of cancer immunotherapy, its effectiveness is often limited in a significant proportion of patients, highlighting the need to explore alternative tumor immune evasion mechanisms. Adenosine, a key metabolite accumulating in hypoxic tumor regions, has emerged as a promising target in oncology. Inhibiting the adenosinergic pathway not only inhibits tumor progression but also holds potential to enhance immunotherapy outcomes. Multiple therapeutic strategies targeting this pathway are being explored, ranging from preclinical studies to clinical trials. This review examines the complex interactions between adenosine, its receptors, and the tumor microenvironment, proposing strategies to target the adenosinergic axis to boost anti-tumor immunity. It also evaluates early clinical data on pharmacological inhibitors of the adenosinergic pathway and discusses future directions for improving clinical responses.
Collapse
Affiliation(s)
- Yuying Yang
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lin Zhu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yantao Xu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Long Liang
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Li Liu
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Hui Li
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Hong Liu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
24
|
Dai P, Sun Y, Huang Z, Liu YT, Gao M, Liu HM, Shi J, He C, Xiang B, Yao Y, Yu H, Xu G, Kong L, Xiao X, Wang X, Zhang X, Xiong W, Hu J, Lin D, Zhong B, Chen G, Gong Y, Xie C, Zhang J. USP2 inhibition unleashes CD47-restrained phagocytosis and enhances anti-tumor immunity. Nat Commun 2025; 16:4564. [PMID: 40379682 PMCID: PMC12084640 DOI: 10.1038/s41467-025-59621-5] [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: 04/01/2024] [Accepted: 04/30/2025] [Indexed: 05/19/2025] Open
Abstract
The CD47/SIRPα axis conveys a 'don't eat me' signal, thereby thwarting the phagocytic clearance of tumor cells. Although blocking antibodies targeting CD47 have demonstrated promising anti-tumor effects in preclinical models, clinical trials involving human cancer patients have not yielded ideal results. Exploring the regulatory mechanisms of CD47 is imperative for devising more efficacious combinational therapies. Here, we report that inhibiting USP2 prompts CD47 degradation and reshapes the tumor microenvironment (TME), thereby enhancing anti-PD-1 immunotherapy. Mechanistically, USP2 interacts with CD47, stabilizing it through deubiquitination. USP2 inhibition destabilizes CD47, thereby boosting macrophage phagocytosis. Single-cell RNA sequencing shows USP2 inhibition reprograms TME, evidenced by increasing M1 macrophages and CD8+ T cells while reducing M2 macrophages. Combining ML364 with anti-PD-1 reduces tumor burden in mouse models. Clinically, low USP2 expression predicts a better response to anti-PD-1 treatment. Our findings uncover the regulatory mechanism of CD47 by USP2 and targeting this axis boosts anti-tumor immunity.
Collapse
Affiliation(s)
- Panpan Dai
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yishuang Sun
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Zhengrong Huang
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu-Tong Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Minling Gao
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Hai-Ming Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jie Shi
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Chuan He
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Bolin Xiang
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Yingmeng Yao
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Haisheng Yu
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Gaoshan Xu
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Lijun Kong
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Xiangling Xiao
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Xiyong Wang
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Xue Zhang
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Wenjun Xiong
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Jing Hu
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Dandan Lin
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bo Zhong
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Yan Gong
- Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Jinfang Zhang
- Department of Radiation and Medical Oncology, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, College of Life Sciences, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
25
|
Li YR, Zhu Y, Halladay T, Yang L. In vivo CAR engineering for immunotherapy. Nat Rev Immunol 2025:10.1038/s41577-025-01174-1. [PMID: 40379910 DOI: 10.1038/s41577-025-01174-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2025] [Indexed: 05/19/2025]
Abstract
Chimeric antigen receptor (CAR)-engineered immune cell therapy represents an important advance in cancer treatments. However, the complex ex vivo cell manufacturing process and stringent patient selection criteria curtail its widespread use. In vivo CAR engineering is emerging as a promising off-the-shelf therapy, providing advantages such as streamlined production, elimination of patient-specific manufacturing, reduced costs and simplified logistics. A large set of preclinical findings has inspired further investigation into treatments for hard-to-treat diseases such as solid tumours and has facilitated the development of advanced products to enhance in vivo CAR engineering efficacy, the persistence of the cellular therapeutic and safety. In this Review, we summarize current in vivo CAR engineering strategies, including nanoparticle-based and viral delivery systems as well as bioinstructive implantable scaffolds, and discuss their advantages and disadvantages. Additionally, we provide a systematic comparison between in vivo and conventional ex vivo CAR engineering methods and address the challenges and future prospects of in vivo CAR engineering.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tyler Halladay
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
26
|
Zang T, Luo X, Mo Y, Lin J, Lu W, Li Z, Zhou Y, Chen S. A novel model for predicting immunotherapy response and prognosis in NSCLC patients. Cancer Cell Int 2025; 25:178. [PMID: 40375214 PMCID: PMC12083170 DOI: 10.1186/s12935-025-03800-3] [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: 01/20/2025] [Accepted: 04/24/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND How to screen beneficiary populations has always been a clinical challenge in the treatment of non-small-cell lung cancer (NSCLC) with immune checkpoint inhibitors (ICIs). Routine blood tests, due to their advantages of being minimally invasive, convenient, and capable of reflecting tumor dynamic changes, have potential value in predicting the efficacy of ICIs treatment. However, there are few models based on routine blood tests to predict the efficacy and prognosis of immunotherapy. METHODS Patients were randomly divided into training cohort and validation cohort at a ratio of 2:1. The random forest algorithm was applied to select important variables based on routine blood tests, and a random forest (RF) model was constructed to predict the efficacy and prognosis of ICIs treatment. For efficacy prediction, we assessed receiver operating characteristic (ROC) curves, decision curve analysis (DCA) curves, clinical impact curve (CIC), integrated discrimination improvement (IDI) and net reclassification improvement (NRI) compared with the Nomogram model. For prognostic evaluation, we utilized the C-index and time-dependent C-index compared with the Nomogram model, Lung Immune Prognostic Index (LIPI) and Systemic Inflammatory Score (SIS). Patients were classified into high-risk and low-risk groups based on RF model, then the Kaplan-Meier (K-M) curve was used to analyze the differences in progression-free survival (PFS) and overall survival (OS) of patients between the two groups. RESULTS The RF model incorporated RDW-SD, MCV, PDW, CD3+CD8+, APTT, P-LCR, Ca, MPV, CD4+/CD8+ ratio, and AST. In the training and validation cohorts, the RF model exhibited an AUC of 1.000 and 0.864, and sensitivity/specificity of (100.0%, 100.0%) and (70.3%, 93.5%), respectively, which had superior performance compared to the Nomogram model (training cohort: AUC = 0.531, validation cohort: AUC = 0.552). The C-index of the RF model was 0.803 in the training cohort and 0.712 in the validation cohort, which was significantly higher than Nomogram model, LIPI and SIS. K-M survival curves revealed that patients in the high-risk group had significantly shorter PFS/OS than those in the low-risk group. CONCLUSIONS In this study, we developed a novel model (RF model) to predict the response to immunotherapy and prognosis in NSCLC patients. The RF model demonstrated better predictive performance for immunotherapy responses than the Nomogram model. Moreover, when predicting the prognosis of immunotherapy, it outperformed the Nomogram model, LIPI, and SIS.
Collapse
Affiliation(s)
- Ting Zang
- The First Clinical Medical College and the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Xiaorong Luo
- The First Clinical Medical College and the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Yangyu Mo
- The First Clinical Medical College and the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Jietao Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
- Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510470, Guangdong, People's Republic of China
| | - Weiguo Lu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Zhiling Li
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Yingchun Zhou
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China.
- Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510470, Guangdong, People's Republic of China.
| | - Shulin Chen
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
27
|
Kim SY, Yi JM, Chun J, Park M, Yeo H, Park SM, Jeong MK. Deciphering the immunomodulatory mechanisms of Bojungikki-tang via systematic transcriptomic and immune cell interaction network analysis. Biomed Pharmacother 2025; 188:118129. [PMID: 40378772 DOI: 10.1016/j.biopha.2025.118129] [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: 03/04/2025] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
Bojungikki-tang (BJIKT), a traditional herbal formula with immunomodulatory properties, has synergistic effects with immune checkpoint inhibitors. However, the detailed molecular mechanisms underlying its effects on various immune cell types remain largely unexplored. Therefore, in this study, we aimed to propose a framework for understanding how herbal medicine modulates immunity through a systematic analysis of the drug-induced transcriptome and immune cell interaction networks. We obtained large-scale RNA-seq data for distinct five immune cell types (T cells, natural killer cells, B cells, macrophages, and dendritic cells) treated with BJIKT and its four major constituent herbs, totaling 180 sequenced samples. Transcriptomic analysis indicated that BJIKT significantly upregulated interferon-γ, TNF-α, and inflammatory pathways in B cells, macrophages, and dendritic cells. Although BJIKT did not directly activate T cells, it indirectly modulated their function through immune cell-cell interactions. The reconstructed network and cytokine assays identified IL-1β, IL-6, IL-8, MIP-1β, CXCL9, CXCL10, and TNF-α as critical cytokines influenced by BJIKT, playing pivotal roles in activating and recruiting various immune cells. Our findings provide insights into the immunomodulatory mechanisms of BJIKT and the unique actions of its constituent herbs. We highlight the potential for combining traditional herbal medicine with omics technologies to explore the therapeutic mechanisms of natural products as complementary therapies for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Sang-Yun Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jin-Mu Yi
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Jaemoo Chun
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Musun Park
- KM Data Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Heerim Yeo
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Sang-Min Park
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea.
| | - Mi-Kyung Jeong
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| |
Collapse
|
28
|
Zhou Y, Kou J, Li W, Wang Y, Su X, Zhang H. BCAA metabolism in cancer progression and therapy resistance: The balance between fuel and cell signaling. Front Pharmacol 2025; 16:1595176. [PMID: 40438606 PMCID: PMC12116492 DOI: 10.3389/fphar.2025.1595176] [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: 03/17/2025] [Accepted: 05/01/2025] [Indexed: 06/01/2025] Open
Abstract
Branched-chain amino acids (BCAAs), including leucine, isoleucine, and valine, play a crucial role in cellular metabolism and signaling. Recent studies have demonstrated that BCAA metabolic reprogramming is a key driver of tumor progression and treatment resistance in various cancers. BCAA metabolism supports cancer cell growth, survival, and proliferation by modulating pathways such as mTOR signaling and oxidative stress responses. By promoting immunosuppressive conditions and increasing the survival rate of cancer stem cells (CSCs), BCAAs contribute to immune evasion and resistance to therapies such as chemotherapy and immune checkpoint inhibitors. This article explores the different metabolic reprogramming patterns of BCAAs in various tumors and introduces BCAA-related metabolic targets for overcoming tumor resistance, offering new directions for precision cancer treatment, reducing resistance, and improving patient outcomes.
Collapse
Affiliation(s)
- Yi Zhou
- Departments of Thoracic Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Jiahui Kou
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Wenjin Li
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Yuyao Wang
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| | - Xingxing Su
- Shunyi Maternal and Children’s Hospital of Beijing Children’s Hospital, Beijing, China
| | - Hongguang Zhang
- Departments of Thoracic Surgery, First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
29
|
Yang F, Xue H, Fan Y, Zhang T, Wang T, Gu F, Guan L, Zhou L, Guan X, Chen G. Engineered hybrid cell membrane nanovesicles for potentiated cancer immunotherapy through dual immune checkpoint inhibition. Biomater Sci 2025; 13:2642-2650. [PMID: 40202456 DOI: 10.1039/d5bm00298b] [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: 04/10/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated remarkable success in treating various types of solid tumors; however, only a limited number of patients currently benefit from these therapeutic agents. Developing novel ICIs that elicit systemic and durable antitumor immune responses remains a significant challenge in improving immunotherapy outcomes. In this study, we engineered PD-1/LAG-3 receptors onto cell membrane nanovesicles to simultaneously block two immune checkpoints for the treatment of colorectal cancer. This dual-checkpoint blockade strategy led to significantly more potent tumor growth suppression in mice with MC38 xenografts compared to nanovesicles targeting PD-1 or LAG-3 alone. Notably, the hybrid nanovesicles substantially rejuvenated exhausted CD8+ T cells, promoting dendritic cell maturation and depleting regulatory T cells (Tregs). This research highlights the promising potential of cell membrane nanovesicles as an effective platform for delivering multiple immune checkpoints in cancer immunotherapy, offering a novel strategy to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Fuxu Yang
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Han Xue
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Yuxin Fan
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Ting Zhang
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
| | - Ting Wang
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
| | - Fanlin Gu
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Longxue Guan
- College of Medical Technology, Beihua University, Jilin 132013, PR China
| | - Lisha Zhou
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
| | - Xingang Guan
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
| | - Guofu Chen
- The First People's Hospital of Wenling (Taizhou University Affiliated Wenling Hospital), School of Medicine, Taizhou University, Taizhou 317500, PR China.
| |
Collapse
|
30
|
Wang S, Zhang D, Ge M, Zhang N, Yang W, Liu Y. Fatty acid metabolism-related risk signature revealing the immune landscape of neuroblastoma and predicting overall survival in pediatric neuroblastoma patients. Discov Oncol 2025; 16:748. [PMID: 40358884 PMCID: PMC12075754 DOI: 10.1007/s12672-025-02479-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Tumor metabolic reprogramming is a hallmark in cancer cells, wherein fatty acid metabolism assumes a pivotal role in energy supply and the provision of diverse biosynthetic precursors. However, there is a lack of systematic analysis regarding the impact of fatty acid metabolism on prognosis in neuroblastoma (NB) patients and its influence on the immune microenvironment. METHODS We acquired RNA expression profiles and corresponding clinical-pathological information for NB patients from the Gene Expression Omnibus, ArrayExpress, and TARGET databases. The GSE49710 cohort was utilized as a training set, whereas E-MTAB-8248 and the TARGET cohorts served as testing sets. Consensus clustering was employed to identify molecular subtypes based on fatty acid metabolism. Independent prognostic genes were pinpointed using LASSO-Cox analysis, which facilitated the development of a novel risk signature that was subsequently validated using the testing sets. We then proceeded to analyze the predictive power of the risk signature for prognosis, its correlation with clinical-pathological features, the immune landscape, and drug sensitivity. RESULTS In the consensus clustering analysis, patients in the training set were segregated into two clusters. Cluster 2 exhibiting significantly poorer overall survival (OS) compared to cluster 1. Moreover, cluster 2 was markedly associated with clinical-pathological features indicative of poor prognosis. Following this, univariate Cox regression analysis revealed 207 fatty acid metabolism genes (FMGs) correlated with patient OS. A risk signature based on 35 FMGs was constructed using LASSO-Cox regression analysis, demonstrating significant predictive accuracy and discrimination in both the training and testing sets. The risk signature emerged as an independent prognostic factor and was integrated with multiple clinical-pathological features to develop a nomogram. In the immune landscape analysis, the high-risk group displayed a compromised antigen presentation mechanism, reduced infiltration levels of various immune cells, and escaping of CD8 + T cells and NK cells. Additionally, different risk groups could exhibit different responsiveness to immune checkpoint inhibitors. Lastly, potential chemotherapeutic agents for each risk group were predicted. CONCLUSION The novel risk signature, derived from FMGs, demonstrated promising efficacy in predicting the prognosis of NB patients, elucidating their immune landscape, and guiding therapeutic strategies.
Collapse
Affiliation(s)
- Shizun Wang
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, China
| | - Dan Zhang
- Beijing Cygenta BioTechnology Co., Ltd, Beijing, China
| | - Ming Ge
- Department of Neurosurgery, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Nijia Zhang
- Department of Neurosurgery, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Wei Yang
- Department of Neurosurgery, National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yuqin Liu
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences (CAMS) and School of Basic Medicine, Peking Union Medical College (PUMC), Beijing, China.
| |
Collapse
|
31
|
Tan AJ, Liu WY, Lu JL, Tan QY, Yan Y, Mo DC. A pharmacovigilance analysis of post-marketing safety of durvalumab. Sci Rep 2025; 15:16661. [PMID: 40360595 PMCID: PMC12075497 DOI: 10.1038/s41598-025-01583-1] [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: 11/22/2024] [Accepted: 05/07/2025] [Indexed: 05/15/2025] Open
Abstract
Durvalumab has demonstrated significant efficacy in several types of malignancies, while large-scale real-world safety studies remain limited. This study aimed to systematically evaluate the safety of durvalumab through data mining of the U.S. Food and Drug Administration Adverse Event Reporting System (FAERS). We extracted reports of durvalumab as the primary suspected drug from the FAERS database (January 2017 to June 2024). Four disproportionality analysis algorithms were used to detect signals between durvalumab and adverse events (AEs). Durvalumab was recorded in 10,120 reports as the primary suspected drug. Of these, 43.6% of AEs occurred during the first month of treatment, with a median onset time of 40 days (IQR: 14-99 ). Among 181 potential signals, 64 were unexpected preferred terms not listed in the prescribing information, including cytokine release syndrome (CRS), pulmonary tuberculosis, radiation esophagitis, oesophageal fistula, oesophageal perforation, pleural effusion, pneumothorax, cerebral infarction, biliary tract infection, cholecystitis, psoriasiform dermatitis, portal vein thrombosis, acute cholangitis and pericarditis malignant. Serious adverse events accounted for 93.3% of cases. Males exhibited a significantly higher risk of experiencing serious outcomes compared to females (OR = 1.83, 95% CI: 1.52-2.19, P < 0.001). Older age groups demonstrated an elevated risk of severe outcomes relative to those under 65 years (65-74 years: OR = 1.52, 95% CI: 1.15-2.00, P = 0.003; ≥75 years: OR = 1.40, 95% CI: 1.02-1.92, P = 0.038). This study comprehensively assessed the safety of durvalumab and discovered potential new adverse event signals, which may provide critical support for risk identification and monitoring of durvalumab.
Collapse
Affiliation(s)
- An-Ju Tan
- Office of Drug Clinical Trials Institutions, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Wan-Ying Liu
- Office of Drug Clinical Trials Institutions, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jun-Li Lu
- Office of Drug Clinical Trials Institutions, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qing-Ying Tan
- Reproductive Medical Center, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yu Yan
- Office of Drug Clinical Trials Institutions, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Dun-Chang Mo
- Department of Tumor Radiotherapy, The Third Affiliated Hospital of Guangxi Medical University, Dan-Cun Road No.13, Nanning, Guangxi, China.
| |
Collapse
|
32
|
Vázquez-Marmolejo AV, Duarte-Mata DI, Ruiz-De La Cruz ML, López-López N, Salinas-Carmona MC, Mejía-Torres M. Increased expression of the PD-1/PD-L1 regulatory axis in tissue leukocytes from experimental actinomycetoma by Nocardia brasiliensis in BALB/c mice. Microb Pathog 2025; 205:107697. [PMID: 40368069 DOI: 10.1016/j.micpath.2025.107697] [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: 11/01/2024] [Revised: 05/08/2025] [Accepted: 05/12/2025] [Indexed: 05/16/2025]
Abstract
The development of experimental actinomycetoma in mice pioneered the study of infective mechanisms in nocardiosis. However, the understanding of lymphocyte activation in actinomycetoma remains incomplete. In this study, we used flow cytometry to evaluate the cellularity and expression of regulatory receptors on leukocytes from spleen and infected tissue in a mice model of experimental actinomycetoma induced by Nocardia brasiliensis. Our results indicate that neutrophils dominate cellularity in infected tissue, representing >90% of infiltrated leukocytes. Among lymphocytes, the percentage of Th1 and Tc1 cells decreases in spleen and infected tissue during chronic infection. Likewise, both tissues had similar changes in leukocyte expression of Programmed Cell Death Protein 1 (PD-1), T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), and Programmed death-ligand 1 (PD-L1), suggesting the delayed systemic involvement of an initially local disease.
Collapse
Affiliation(s)
- Anna Velia Vázquez-Marmolejo
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México
| | - Diana Ivonne Duarte-Mata
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México
| | - María Luisa Ruiz-De La Cruz
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México
| | - Nallely López-López
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México
| | - Mario César Salinas-Carmona
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México
| | - Manuel Mejía-Torres
- Universidad Autónoma de Nuevo León, Service of Immunology, Hospital Universitario "Dr. José Eleuterio González", Monterrey, NL, México.
| |
Collapse
|
33
|
Yao J, Gan W, Sun J, Han Z, Li D, Cao L, Zhu L. APOL6 as a potential biomarker of immuno-correlation and therapeutic prediction in cancer immunotherapy. Medicine (Baltimore) 2025; 104:e42406. [PMID: 40355224 PMCID: PMC12073870 DOI: 10.1097/md.0000000000042406] [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: 04/23/2024] [Accepted: 04/22/2025] [Indexed: 05/14/2025] Open
Abstract
The emergence of immune checkpoint inhibitors (ICIs) has significantly revolutionized the approach to treating advanced cancers. Despite their remarkable efficacy, not all patients exhibit favorable responses to ICI therapy. Hence, more biomarkers for therapeutic prediction need to be discovered. In this study, we utilized public cohorts to investigate the predictive significance and immunological associations of apolipoprotein L6 (APOL6) in cancers. The expression of APOL6 was found to be enhanced in tumors of patients who exhibited strong immunotherapeutic responses across various types of cancer. Furthermore, APOL6 showed immune correlations in pan-cancer and was confirmed by the tissue microarray cohort and in vitro experiments. Overall, this study highlights that APOL6 serves as a beneficial biomarker for immune checkpoint inhibitors in patients with cancer. Additional research involving larger numbers of patients and the underlying mechanism is necessary to determine its effectiveness as a biomarker for predicting the benefits of ICIs.
Collapse
Affiliation(s)
- Jialin Yao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenyuan Gan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiukang Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhihang Han
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dongqing Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Cao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Oncology, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
34
|
Jiang D, Yoou MS, Cho S, Choi Y. Molecular Dynamics-Guided Repositioning of FDA-Approved Drugs for PD-L1 Inhibition with In Vitro Anticancer Potential. Int J Mol Sci 2025; 26:4497. [PMID: 40429641 PMCID: PMC12110937 DOI: 10.3390/ijms26104497] [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: 04/03/2025] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Programmed death-ligand 1 (PD-L1) is a crucial immune checkpoint protein that tumors often exploit to evade immune surveillance. This study systematically screened a library of 1031 FDA-approved drugs using a high-throughput molecular dynamics approach to identify potential inhibitors targeting PD-L1. From this screening, five promising compounds-vorapaxar, delafloxacin, tenofovir disoproxil, pivmecillinam, and fursultiamine-showed significant binding affinities to PD-L1 and demonstrated cytotoxic activity against A549 lung tumor cells. These candidates were further evaluated through extended molecular dynamics simulations lasting up to 150 ns to assess their structural stability, residue fluctuations, and binding free energy. Among the identified compounds, pivmecillinam demonstrated the most favorable results, exhibiting stable binding interactions and a binding free energy of -18.01 kcal/mol, comparable to that of the known PD-L1 inhibitor BMS-1. These findings suggest that pivmecillinam has promising immunomodulatory potential and could serve as a candidate for further development in cancer immunotherapy. Overall, this study underscores the value of integrating high-throughput MD and experimental approaches for drug repositioning to identify novel therapeutic agents.
Collapse
Affiliation(s)
- Dejun Jiang
- Department of Environmental Engineering, Hoseo University, Asan 31499, Republic of Korea;
| | - Myoung-Schook Yoou
- Eulji Medi-Bio Research Institute, Eulji University, Daejeon 34824, Republic of Korea;
| | - Sungjoon Cho
- Department of Bio-Applied Toxicology, Hoseo University, Asan 31499, Republic of Korea;
| | - Youngjin Choi
- Department of Bio-Applied Toxicology, Hoseo University, Asan 31499, Republic of Korea;
- Department of Food Science & Technology, Hoseo University, Asan 31499, Republic of Korea
| |
Collapse
|
35
|
Chu X, Pu N, Yang X, Xie Y, Liu L, Jin Y. Subtypes of tumor-associated neutrophils and their roles in cancer immunotherapy. Crit Rev Oncol Hematol 2025; 212:104763. [PMID: 40334802 DOI: 10.1016/j.critrevonc.2025.104763] [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: 03/10/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025] Open
Abstract
Neutrophils are essential components of the innate immune system. Tumor-associated neutrophils (TANs) are shaped by tumor microenvironment (TME), leading to significant heterogeneity in biological characteristics and functions. Recent advances in single-cell sequencing have revealed a wide array of TAN subtypes, while a comprehensive classification system is still lacking. This review aims to summarize the alterations observed in TAN subgroups following cancer immunotherapy, and identify the distinctions and commonalities between pro-tumor and anti-tumor subgroups. Current progress of preclinical and clinical studies is also highlighted, involving novel therapies targeting TANs.
Collapse
Affiliation(s)
- Xinyun Chu
- Department of Hepatobiliary & Pancreatic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650000, China; Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ning Pu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xue Yang
- Department of Hematology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuqi Xie
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Yun Jin
- Department of Hepatobiliary & Pancreatic Surgery, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650000, China.
| |
Collapse
|
36
|
Maruyama Y, Hosonuma M, Toyoda H, Funayama E, Sasaki A, Baba Y, Tajima K, Nakashima R, Sasaki A, Shida M, Tsurui T, Tsuji M, Tsunoda T, Shimane T, Kiuchi Y, Yoshimura K, Kuramasu A. Combination therapy with cetirizine and anti-PD-1 antibody suppresses colitis-induced colon tumor formation in mice. Eur J Pharmacol 2025; 999:177704. [PMID: 40320111 DOI: 10.1016/j.ejphar.2025.177704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 04/24/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have transformed cancer therapy, yet their efficacy remains limited in inflammation-associated tumors, necessitating combinatorial approaches. Although combinations with cytotoxic or targeted agents are well established, the therapeutic potential of non-oncologic drugs, such as antihistamines, is less explored. In this study, we investigate whether cetirizine, a non-sedating histamine H1 receptor antagonist, enhances the antitumor effects of anti-programmed cell death protein 1 (PD-1) antibody in a murine model of colitis-associated colorectal cancer. Combination therapy, not monotherapy, significantly reduced tumor volume in vivo. Flow cytometry of splenocytes revealed increased PD-1 expression on T cells only in the combination group, suggesting systemic immune activation. Immunohistochemical analysis showed elevated CD3+ T-cell infiltration into tumors following combination treatment. Meanwhile, gene expression analysis of tumor tissues revealed downregulated Vegfa, Mmp9, Il10, and Cd80, along with upregulated Hspg2 and Fn1, suggesting a shift in the tumor microenvironment. In vitro, cetirizine suppressed Mmp9 expression in CT26 cells, Il10 in macrophages, and VEGFA in human umbilical vein endothelial cells, indicating cell-type-specific effects that partially mirror the in vivo findings. Immunohistochemistry further demonstrated a reduced frequency of FoxP3+ regulatory T cells among CD3+ T cells within the tumor stroma in the combination group. Collectively, these findings indicate that cetirizine enhances ICI efficacy by reshaping the tumor microenvironment through immunomodulatory mechanisms. Our results support the repurposing of antihistamines as a novel strategy to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Yuki Maruyama
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan; Department of Otorhinolaryngology-Head and Neck Surgery, Showa Medical University School of Medicine, Tokyo, Japan
| | - Masahiro Hosonuma
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan; Division of Medical Oncology, Department of Medicine, Showa Medical University School of Medicine, Tokyo, Japan
| | - Hitoshi Toyoda
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan; Department of Orthopaedic Surgery, School of Medicine, Showa Medical University, Tokyo, Japan
| | - Eiji Funayama
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan; Division of Pharmacology, Department of Pharmacology, Toxicology and Therapeutics, School of Pharmacy, Showa Medical University, Tokyo, Japan
| | - Akiko Sasaki
- Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan
| | - Yuta Baba
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan
| | - Kohei Tajima
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan
| | - Rie Nakashima
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan
| | - Aya Sasaki
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan
| | - Midori Shida
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan
| | - Toshiaki Tsurui
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan; Division of Medical Oncology, Department of Medicine, Showa Medical University School of Medicine, Tokyo, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa Medical University, Tokyo, Japan
| | - Takuya Tsunoda
- Division of Medical Oncology, Department of Medicine, Showa Medical University School of Medicine, Tokyo, Japan
| | - Toshikazu Shimane
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Head and Neck Oncology Center, Showa Medical University, Tokyo, Japan
| | - Yuji Kiuchi
- Division of Medical Pharmacology, Department of Pharmacology, Showa Medical University School of Medicine, Tokyo, Japan; Pharmacological Research Center, Showa Medical University, Tokyo, Japan
| | - Kiyoshi Yoshimura
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan; Division of Medical Oncology, Department of Medicine, Showa Medical University School of Medicine, Tokyo, Japan
| | - Atsuo Kuramasu
- Department of Clinical Immuno Oncology, Research Institute for Clinical Pharmacology and Therapeutics, Showa Medical University, Tokyo, Japan.
| |
Collapse
|
37
|
Qu WF, Zhu GQ, Yang R, Chu TH, Guan ZQ, Huang R, Tian MX, Jiang XF, Tao CY, Fang Y, Gao J, Wu XL, Chen JF, Zhao QF, Wang Y, Bu YC, Zhou J, Fan J, Liu WR, Tang Z, Shi YH. Targeting HMGB2 acts as dual immunomodulator by bolstering CD8 + T cell function and inhibiting tumor growth in hepatocellular carcinoma. SCIENCE ADVANCES 2025; 11:eads8597. [PMID: 40315321 PMCID: PMC12047442 DOI: 10.1126/sciadv.ads8597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 03/27/2025] [Indexed: 05/04/2025]
Abstract
T cell exhaustion is a critical obstacle for durable treatment response in hepatocellular carcinoma (HCC). Developing drugs that control tumor growth and simultaneously bolster immune function is of great significance. Although high-mobility group box 2 (HMGB2) has been reported to be crucial to HCC prognosis, its role in the tumor microenvironment remains unclear. Here, we found HMGB2+ CD8+ T cells as being associated with immune exhaustion and resistance to anti-PD-1 treatment through single-cell RNA sequencing. Mechanistically, HMGB2 impaired the oxidative phosphorylation in CD8+ T cells and inactivated the interferon-γ response in tumor cells, reducing the antitumor effector function. Tannic acid, a specific inhibitor of HMGB2, synergized with PD-1 antibody to attenuate tumor growth and reverse T cell exhaustion. Our findings highlight the unique role of HMGB2 as an immune exhaustion associated molecule. Targeting HMGB2 on both CD8+ T cells and tumor cells contributed to promising treatment strategies for HCC.
Collapse
Affiliation(s)
- Wei-Feng Qu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
- Department of Thyroid and Breast Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Gui-Qi Zhu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Yang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Tian-Hao Chu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhi-Qi Guan
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Run Huang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
- Department of Thyroid and Breast Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meng-Xin Tian
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xi-Fei Jiang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Chen-Yang Tao
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuan Fang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Jun Gao
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao-Ling Wu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Jia-Feng Chen
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Qian-Fu Zhao
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Yi Wang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Yi-Chao Bu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Jian Zhou
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Jia Fan
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei-Ren Liu
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Zheng Tang
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| | - Ying-Hong Shi
- Department of Liver Surgery, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Research Unit of Liver Cancer Recurrence and Metastasis, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
38
|
Tayebi-Khorrami V, Fadaei MR, Fallahianshafiei S, Askari VR. Immune checkpoint blocking in cancer therapy using thermosensitive hydrogels: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04171-2. [PMID: 40314764 DOI: 10.1007/s00210-025-04171-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/09/2025] [Indexed: 05/03/2025]
Abstract
Cancer is a challenging issue requiring new strategies for management and control. Immune checkpoint blockades (ICBs) increase the body's immune response against cancer by targeting specific receptors on T-lymphocytes. The FDA approved different ICBs for cancer treatment: anti-PD-1, PDL-1, and CTLA-4 inhibitors. Many immune checkpoint inhibitors (ICIs) are in clinical trials, highlighting their significance. Challenges like resistance and side effects have led researchers to explore new delivery strategies for ICIs. Thermosensitive hydrogels can change from sol to gel and vice versa due to their structure. They interact with aqueous medium through groups like ethyl, methyl, and propyl, forming hydrogen bonds. These bonds of hydrogen are temperature-sensitive and cause the change of the polymer from sol to gel at a temperature named critical solution temperature (CST). The using temperature-responsive polymers and ICBs showed a promising approach to sustained localized cancer therapy with lowering side effects on normal tissues. In this paper, we first define new investigations on immune therapy in cancer via ICBs. Then, we present recent studies of thermosensitive polymers in cancer therapy and the most used thermosensitive polymers in studies. Eventually, we discuss studies that used thermosensitive polymers in the delivery of ICBs and discuss new investigations in this field.
Collapse
Affiliation(s)
- Vahid Tayebi-Khorrami
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Fadaei
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Vahid Reza Askari
- Clinical Research Development Unit, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
39
|
Summer M, Riaz S, Ali S, Noor Q, Ashraf R, Khan RRM. Understanding the Dual Role of Macrophages in Tumor Growth and Therapy: A Mechanistic Review. Chem Biodivers 2025; 22:e202402976. [PMID: 39869825 DOI: 10.1002/cbdv.202402976] [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: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
Macrophages are heterogeneous cells that are the mediators of tissue homeostasis. These immune cells originated from monocytes and are classified into two basic categories, M1 and M2 macrophages. M1 macrophages exhibit anti-tumorous inflammatory reactions due to the behavior of phagocytosis. M2 macrophages or tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) and have a basic role in tumor progression by interacting with other immune cells in TME. By the expression of various cytokines, chemokines, and growth factors, TAMs lead to strengthening tumor cell proliferation, angiogenesis, and suppression of the immune system which further support invasion and metastasis. This review discusses recent and updated mechanisms regarding tumor progression by M2 macrophages. Moreover, the current therapeutic approaches targeting TAMs, their advantages, and limitations are also summarized, and further treatment approaches are outlined along with an elaboration of the tumor regression role of macrophages. This comprehensive review article possibly helps to understand the mechanisms underlying the tumor progression and regression role of macrophages in a comparative way from a basic level to the advanced one.
Collapse
Affiliation(s)
- Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Saima Riaz
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Qudsia Noor
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rimsha Ashraf
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, Government College University, Lahore, Pakistan
| | - Rana Rashad Mahmood Khan
- Faculty of Chemistry and Life Sciences, Department of Chemistry, Government College University Lahore, Lahore, Pakistan
| |
Collapse
|
40
|
Wang H, Baba Y, Hara Y, Toihata T, Kosumi K, Harada K, Iwatsuki M, Miyamoto Y, Baba H. The Relationship Between Gut Microbiome Bifidobacterium and Anti-tumor Immune Responses in Esophageal Squamous Cell Carcinoma. Ann Surg Oncol 2025; 32:3828-3838. [PMID: 40035906 PMCID: PMC11976794 DOI: 10.1245/s10434-024-16288-4] [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: 07/10/2024] [Accepted: 09/17/2024] [Indexed: 03/06/2025]
Abstract
BACKGROUND The Bifidobacterium genus is a prominent bacterial population in the gastrointestinal tract. Previous findings suggest that Bifidobacterium is linked to tumor suppression in mouse models of melanoma. Additionally, when combined with the programmed death-ligand 1 (PD-L1) antibody, it can enhance anti-tumor treatment by increasing tumor-specific T-cell responses and promoting infiltration of antigen-specific T cells into tumors. However, there is a lack of studies on Bifidobacterium in esophageal squamous cell carcinoma (ESCC). This study aimed to investigate the potential impact of Bifidobacterium on this cancer type. METHODS We examined 213 samples from ESCC patients who underwent tumor resection. The presence of Bifidobacterium was confirmed using quantitative polymerase chain reaction and fluorescent in situ hybridization (FISH). Patient overall survival (OS) was analyzed with Bifidobacterium positivity. Tumor-infiltrating lymphocytes (TILs) were evaluated via hematoxylin and eosin stains, and immunohistochemistry was used to assess programmed death-1 (PD-1), PD-L1, cluster of differentiation 8 (CD8), and forkhead box P3 (FOXP3) expression. Nutritional status was evaluated via computed tomography scans. RESULTS Bifidobacterium positivity showed no correlation with patient OS or TIL levels; however, Bifidobacterium positivity in normal tissue was associated with lower FOXP3 levels, suggesting a potential role in upregulating anti-tumor immune responses. Patients with Bifidobacterium present in peritumor normal tissue exhibited better skeletal muscle area and volume. Conversely, Bifidobacterium positivity in tumor tissue was associated with poorer prognostic nutrition index values, likely due to decreased albumin levels. CONCLUSION Bifidobacterium can induce the upregulated anti-tumor immune response and is more prevalent in cases with good nutritional status.
Collapse
Affiliation(s)
- Haolin Wang
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Yoshihiro Hara
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tasuku Toihata
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keisuke Kosumi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuto Harada
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
41
|
van Dijk B, Janssen JC, van Daele PLA, de Jonge MJA, Joosse A, Verheul HMW, Epker JL, van der Veldt AAM. From ICI to ICU: A systematic review of patients with solid tumors who are treated with immune checkpoint inhibitors (ICI) and admitted to the intensive care unit (ICU). Cancer Treat Rev 2025; 136:102936. [PMID: 40222269 DOI: 10.1016/j.ctrv.2025.102936] [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: 02/18/2025] [Revised: 04/04/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) have improved the survival of patients with different solid tumors and even resulted in cure of metastatic disease. Since the introduction of ICIs, an increasing number of patients is admitted to the ICU for severe and potentially life-threatening immune related adverse events (irAEs). The outcome of patients who are admitted to the ICU because of severe irAEs is still unknown. The aim of this systematic review is to collect evidence on the outcomes of patients with solid tumors who are admitted to the ICU because of irAEs. METHODS Medline, Embase, Cochrane central register of controlled trials and Google Scholar were searched systematically from 1975 to 24 September 2024. Articles were only included when describing patients with solid tumors who were admitted to the ICU because of irAEs after treatment with ICIs. Two independent reviewers extracted the data and assessed the risk of bias. RESULTS A total of 183 articles were included: two prospective ICU population-based studies, four retrospective ICU population-based studies, 25 retrospective studies describing irAEs with incidental ICU admissions, one review of case reports, and 153 articles with a total of 177 case reports. The six ICU population-based studies contained a total of 169 patients who were admitted to the ICU due to irAEs. In these six studies, the most frequently reported irAEs were pneumonitis and neurological irAEs. Of these 169 patients, 26% of the patients died on the ICU and an additional 8% of patients in the three to six months thereafter due to irAEs or disease progression. In all 183 included articles, various irAEs were described and the reported mortality rate varied from 0 to 53%. CONCLUSION The potential favorable outcomes of both the solid tumors and irAEs will probably result in more need for ICU admissions. Prospective clinical trials are needed to optimize the treatment strategy of severe irAEs at the ICU. Based on the favourable outcomes after life-threatening irAEs, ICU admission should definitely be considered for patients with solid tumors who have life-threatening irAEs.
Collapse
Affiliation(s)
- Brigit van Dijk
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Joséphine C Janssen
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands; Department of Oncological Surgery, Erasmus MC, Rotterdam, the Netherlands
| | - Paul L A van Daele
- Department of Clinical Immunology, Erasmus MC, Rotterdam, the Netherlands
| | - Maja J A de Jonge
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Arjen Joosse
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Henk M W Verheul
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Jelle L Epker
- Department of Intensive Care, Erasmus MC, Rotterdam, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands.
| |
Collapse
|
42
|
Huang M, Zhang Y, Chen Z, Yu X, Luo S, Peng X, Li X. Gut microbiota reshapes the TNBC immune microenvironment: Emerging immunotherapeutic strategies. Pharmacol Res 2025; 215:107726. [PMID: 40184763 DOI: 10.1016/j.phrs.2025.107726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with limited treatment options and poor prognosis. The gut microbiota, a diverse community of microorganisms in the gastrointestinal tract, plays a crucial role in regulating immune responses through the gut-immune axis. Recent studies have highlighted its significant impact on TNBC progression and the efficacy of immunotherapies. This review examines the interactions between gut microbiota and the immune system in TNBC, focusing on key immune cells and pathways involved in tumor immunity. It also explores microbiota modulation strategies, including probiotics, prebiotics, dietary interventions, and fecal microbiota transplantation, as potential methods to enhance immunotherapeutic outcomes. Understanding these mechanisms offers promising avenues for improving treatment efficacy and patient prognosis in TNBC.
Collapse
Affiliation(s)
- Mingyao Huang
- School of Basic Medicine, Putian University, Putian, Fujian 351100, China
| | - Yikai Zhang
- School of Basic Medicine, Putian University, Putian, Fujian 351100, China
| | - Zhaoji Chen
- School of Basic Medicine, Putian University, Putian, Fujian 351100, China
| | - Xin Yu
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian 350011, China
| | - Shiping Luo
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, Fujian 350011, China.
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China; Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management, China.
| | - Xuexin Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Institute of Health Sciences, China Medical University, Shenyang, Liaoning 110122, China; Department of Physiologyand Pharmacology, Karolinska Institutet, Solna 171 65, Sweden.
| |
Collapse
|
43
|
Yu J, Li Y, Yang Y, Guo H, Chen Y, Yi P. PD-1 inhibitors improve the efficacy of tyrosine kinase inhibitors combined with transcatheter arterial chemoembolization in advanced hepatocellular carcinoma: a meta-analysis and trial sequential analysis. Scand J Gastroenterol 2025; 60:472-484. [PMID: 40152031 DOI: 10.1080/00365521.2025.2479193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/21/2025] [Accepted: 03/09/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND This meta-analysis and trial sequential analysis (TSA) aimed to evaluate the efficacy and safety of triple therapy with tyrosine kinase inhibitors (TKIs) combined with transcatheter arterial chemoembolization (TACE) plus programmed death 1 (PD-1) inhibitors (T-T-P) and dual therapy with TKIs combined with TACE (T-T) for the treatment of advanced unresectable hepatocellular carcinoma (uHCC). METHODS Literature related to the efficacy of TKIs combined with TACE plus PD-1 inhibitors in uHCC was searched using the Embase, PubMed, and Cocrane libraries. TSA was used to reduce false positive results due to random error. RESULTS Seventeen articles were included in this meta-analysis, including 2,561 patients. In the T-T-P group, OS [HR 0.45, 95% confidence interval (CI) 0.39-0.52; p = 0.000], PFS [HR 0.43, 95% CI 0.38 - 0.48; p = 0.000], were significantly prolonged compared to those in the T-T group; ORR (RR 1.59 [95% CI 1.39-1.81]; p = 0.000) and DCR (RR 1.26 [95% CI 1.15-1.37]; p = 0.000) were significantly higher. TSA analysis showed early results without further testing. Prognostic factor analysis demonstrated that portal vein tumor thrombus (PVTT) and extrahepatic metastasis were common independent risk factors for OS and PFS. Regarding grade 3/4 adverse events results showed no statistically significant differences in any of them. CONCLUSIONS Compared with T-T treatment group, the T-T-P treatment group exhibited a notable improvement in OS and PFS, particularly in cases of PVTT and extrahepatic metastasis. Furthermore, it can markedly enhance the ORR and DCR in patients with uHCC.
Collapse
Affiliation(s)
- Jiahui Yu
- Department of hepato-biliary-pancrease II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P. R. China
| | - Yong Li
- Department of hepato-biliary-pancrease II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P. R. China
| | - Yuting Yang
- Department of Educational Technology, Institute of Education, China West Normal University, Nanchong, Sichuan, P. R. China
| | - Hao Guo
- Department of hepato-biliary-pancrease II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P. R. China
| | - Yimiao Chen
- Department of hepato-biliary-pancrease II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P. R. China
| | - Pengsheng Yi
- Department of hepato-biliary-pancrease II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, P. R. China
| |
Collapse
|
44
|
Wu M, Liu J, Liu L, Yang Y, Liu H, Yu L, Zeng H, Yuan S, Xu R, Liu H, Jiang H, Qu S, Wang L, Chen Y, Wang J, Zhang Y, He S, Feng L, Han J, Zeng W, Wang H, Huang Y. Autologous Peripheral Vγ9Vδ2 T Cell Synergizes with αβ T Cell Through Antigen Presentation and BTN3A1 Blockade in Immunotherapy of Cervical Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2401230. [PMID: 40091603 PMCID: PMC12079532 DOI: 10.1002/advs.202401230] [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] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/07/2025] [Indexed: 03/19/2025]
Abstract
New treatment strategies are urgently needed for patients with advanced cervical cancer (CC). Here, a synergistic anti-CC effect of a novel combinatorial immunotherapy with adoptively transferred autologous Vγ9Vδ2 T cells and αβ T cells is shown. The pivotal role of both circulating and tumor-infiltrating Vγ9Vδ2 T cells in anti-CC immunity is uncovered. Importantly, autologous Vγ9Vδ2 T cells show a synergistic anti-CC effect with αβ T cells not only through killing tumor directly, but also by promoting the activation and tumoricidal activity of syngeneic αβ T cells through antigen presentation, which can be further boosted by conventional chemotherapy. Moreover, Vγ9Vδ2 T cells can restore the tumoricidal function of αβ T cell through competitively binding to BTN3A1, a TCR-Vγ9Vδ2 ligand on CC cells upregulated by IFN-γ derived from activated αβ T cell. These findings uncover a critical synergistic effect of autologous Vγ9Vδ2 T cells and αβ T cells in immunotherapy of CC and reveal the underlying mechanisms.
Collapse
Affiliation(s)
- Min Wu
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Jian Liu
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Liting Liu
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Yifan Yang
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Hong Liu
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Long Yu
- Beckman Coulter Commercial Enterprise (China) Co., LtdShanghai200122China
| | - Haihong Zeng
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Shuo Yuan
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Ruiyi Xu
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Hangyu Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Han Jiang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Shen Qu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Liming Wang
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineHangzhouZhejiang310006China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Jingyu Wang
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Yuwei Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Shan He
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ling Feng
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Junyan Han
- Department of Immunology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Wanjiang Zeng
- Department of Obstetrics and Gynecology, Tongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Hui Wang
- Department of Obstetrics and GynecologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyCancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Gynecologic Oncology, Women's HospitalZhejiang University School of MedicineZhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science and TechnologyWuhan430030China
| |
Collapse
|
45
|
Cohen RB, Jimeno A, Hreno J, Sun L, Wallén-Öhman M, Millrud CR, Sanfridson A, Garcia-Ribas I. Safety, tolerability, and preliminary efficacy of nadunolimab, an anti-IL- 1 receptor accessory protein monoclonal antibody, in combination with pembrolizumab in patients with solid tumors. Invest New Drugs 2025:10.1007/s10637-025-01538-3. [PMID: 40310569 DOI: 10.1007/s10637-025-01538-3] [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: 03/08/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
Interleukin (IL)-1 signaling has an essential role in tumor progression and immunosuppression and is linked to acquired resistance to anti-PD-1/PD-L1 treatment. Nadunolimab is an IL1RAP (IL-1 receptor accessory protein)-targeting antibody that blocks IL-1α/IL-1β signaling and has enhanced antibody-dependent cellular cytotoxicity. We investigated the safety and preliminary efficacy of nadunolimab with pembrolizumab in patients with metastatic solid tumors who had progressed on previous checkpoint inhibitor treatment, suggesting acquired checkpoint inhibitor resistance (NCT04452214). This phase 1b trial enrolled patients with metastatic disease who had exhausted or declined standard-of-care alternatives. Patients received nadunolimab (5 mg/kg) and standard-dose pembrolizumab. The primary objective was to assess safety. Secondary objectives were anti-tumor response as per iRECIST, pharmacokinetics, and changes in immune mediators. Fifteen patients with stage IV cancer (head and neck squamous cell carcinoma, non-small cell lung cancer, melanoma) entered the trial. Grade ≥ 3 adverse events were reported for 7 patients (47%). There was one dose-limiting toxicity of febrile neutropenia. The most frequent grade ≥ 3 adverse event was dysphagia (two patients). Seven patients (47%) had reductions in target lesion size. Median iPFS was 3.4 months (95% CI 1.4-8.6). Median OS was 19.7 months (95% CI 4.3-28.7) with 67% 1-year survival. Survival was significantly longer in patients with higher baseline tumor infiltration of CD163 + macrophages and natural killer cells and in patients with reduced on-treatment circulating IL-6 levels or neutrophil-to-lymphocyte ratio. Nadunolimab with pembrolizumab had an acceptable safety profile, and prolonged disease control was observed in a subset of patients. The results support further development of nadunolimab in combination with checkpoint inhibitors.
Collapse
Affiliation(s)
- Roger B Cohen
- Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, PA, USA.
| | - Antonio Jimeno
- Department of Medicine, Division of Medical Oncology, Developmental Therapeutics Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jennifer Hreno
- Department of Medicine, Division of Medical Oncology, Developmental Therapeutics Program, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lova Sun
- Division of Hematology-Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
46
|
Tao L, Zhang Y, Zhang J, Tao J, Gong Y, Mao J, Tian Q, Ao P, Zhuo D. Peptides-functionalized gold nanostars enhanced degradation of PD-L1 for improved prostate cancer immunotherapy. J Biomater Appl 2025; 39:1188-1201. [PMID: 39993230 DOI: 10.1177/08853282251319473] [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: 02/26/2025]
Abstract
Blockage of the interaction between programmed death receptor-1 (PD-1) and programmed death ligand-1 (PD-L1) can restore T-cell activity and enhance antitumor immunity. PD-1/PD-L1 pathway inhibitors have promising applications in the treatment of advanced prostate cancer (PCa). We successfully developed a peptides-functionalized gold nanoconstruct (P-AuNS) consisted of PD-L1-binding peptide (PD-L1pep, P) and gold nanostar (AuNS), which could bind to cell-surface PD-L1 specifically and deliver PD-L1 into PCa cells with high efficiency. In PCa cells, P-AuNS can efficiently degrade PD-L1 in a lysosomal-dependent manner. In the co-culture system of Jurkat cells and DU145 cells, P-AuNS restored the proliferative capacity and interferon-gamma (IFN-γ) secretion level of Jurkat cells inhibited by co-cultured DU145 cells, indicating that P-AuNS effectively hampered the interaction between PD-1 and PD-L1. In addition, in PCa-bearing mice, P-AuNS can effectively inhibit tumor growth and down-regulate PD-L1 protein levels, and in vivo experimental results show that P-AuNS has no systemic toxicity. P-AuNS block the interaction between PD-1 and PD-L1 by efficiently degrading PD-L1, thus restoring the antitumor activity of T cells and inhibiting tumor progression of PCa. In all, P-AuNS has great promise as a potential immunotherapy strategy in the treatment of advanced PCa and even other solid tumors.
Collapse
Affiliation(s)
- Liangjun Tao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Yifei Zhang
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Jingwei Zhang
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Jianping Tao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Yu Gong
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Jun Mao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Qixing Tian
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Ping Ao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Dong Zhuo
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, People's Republic of China
| |
Collapse
|
47
|
Erritzøe-Jervild M, Møller SN, Kruuse C, Stenør C. Immune checkpoint inhibitor-related CNS vasculitis - A systematic review and report of 6 cases. J Stroke Cerebrovasc Dis 2025; 34:108265. [PMID: 39984148 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108265] [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: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) represent an important new class of immunotherapy used in cancer treatment. Though effective, immune-related adverse events (irAE) are reported, including cerebral vasculitis (nirVasculitis). In this systematic review, we aim to identify clinical and laboratory features of nirVasculitis and exemplify these in six local clinical cases. OBSERVATIONS We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Two independent researchers searched, identified, and extracted data from both PubMed and Embase to identify reports on nirVasculitis. Based on current criteria for diagnostic certainty patients were categorized as having definite, probable, or possible nirVasculitis. 20 cases described relevant symptomatology and met our inclusion criteria. Non-small-cell lung cancer (55 %) was the most frequent cancer type. Most cases (95 %) received a programmed death-1 (PD-1) inhibitor. One patient had definite vasculitis, seven probable vasculitis, and twelve possible vasculitis. Signs and symptoms included aphasia (n = 5), loss of consciousness (n = 7), confusion (n = 8), unilateral sensory or motor dysfunction (n = 5), and fever or headache (n = 9). All patients had brain imaging and seventeen underwent a lumbar puncture. Of these, 64.7 % had pleocytosis and 52.9 % elevated protein in cerebrospinal fluid. Nineteen patients received corticosteroids. Eight patients fully or partially recovered. Eight patients died due to nirVasculitis, three to advanced cancer, and one lost to follow-up. CONCLUSIONS There are few clinical reports of nirVasculitis which may be due to underreporting or rarity of complications. Guidelines for diagnostics and reporting may improve awareness and early recognition to initiate important immunosuppressive treatment.
Collapse
Affiliation(s)
- Mai Erritzøe-Jervild
- University of Copenhagen, Copenhagen, Denmark; Neurovascular Research Unit (NVRU), Department of Neurology, Copenhagen University Hospital - Herlev and Gentofte Copenhagen, Denmark.
| | | | - Christina Kruuse
- University of Copenhagen, Copenhagen, Denmark; Neurovascular Research Unit (NVRU), Department of Neurology, Copenhagen University Hospital - Herlev and Gentofte Copenhagen, Denmark; Department of Brain- and Spinal Cord Injury, Neuroscience Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Christian Stenør
- University of Copenhagen, Copenhagen, Denmark; Neurovascular Research Unit (NVRU), Department of Neurology, Copenhagen University Hospital - Herlev and Gentofte Copenhagen, Denmark
| |
Collapse
|
48
|
Lindberg A, Muhl L, Yu H, Hellberg L, Artursson R, Friedrich J, Backman M, Hekmati N, Mattsson J, Lindskog C, Brunnström H, Botling J, Mezheyeuski A, Broström E, Gulyas M, Kärre K, Isaksson J, Micke P, Strell C. In Situ Detection of Programmed Cell Death Protein 1 and Programmed Death Ligand 1 Interactions as a Functional Predictor for Response to Immune Checkpoint Inhibition in NSCLC. J Thorac Oncol 2025; 20:625-640. [PMID: 39743139 DOI: 10.1016/j.jtho.2024.12.026] [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: 09/23/2024] [Revised: 12/06/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) have transformed lung cancer treatment, yet their effectiveness seem restricted to certain patient subsets. Current clinical stratification on the basis of programmed death ligand 1 (PD-L1) expression offers limited predictive value. Given the mechanism of action, directly detecting spatial programmed cell death protein 1 (PD1)-PD-L1 interactions might yield more precise insights into immune responses and treatment outcomes. METHODS We applied a second-generation in situ proximity ligation assay to detect PD1-PD-L1 interactions in diagnostic tissue samples from 16 different cancer types, a tissue microarray with surgically resected early-stage NSCLC, and finally diagnostic biopsies from 140 patients with advanced NSCLC with and without ICI treatment. RNA sequencing analysis was used to identify potential resistance mechanisms. RESULTS In the early-stage NSCLC, only approximately half of the cases with detectable PD-L1 and PD1 expression exhibited PD1-PD-L1 interactions, with significantly lower levels in EGFR-mutated tumors. Interaction levels varied across cancer types, aligning with reported ICI response rates. In ICI-treated patients with NSCLC, higher PD1-PD-L1 interactions were linked to complete responses and longer survival, outperforming standard PD-L1 expression assays. Patients who did not respond to ICIs despite high PD1-PD-L1 interactions exhibited additional expression of stromal immune mediators (EOMES, HAVCR1/TIM-1, JAML, FCRL1). CONCLUSION Our study proposes a diagnostic shift from static biomarker quantification to assessing active immune pathways, providing more precise ICI treatment. This functional concept applies to tiny lung biopsies and can be extended to further immune checkpoints. Accordingly, our results indicate concerted ICI resistance mechanisms, highlighting the need for combination diagnostics and therapies.
Collapse
Affiliation(s)
- Amanda Lindberg
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Lars Muhl
- Department of Medicine (Huddinge), Karolinska Institutet, Huddinge, Sweden; Centre of Cancer Biomarkers (CCBIO), Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Hui Yu
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Louise Hellberg
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Rebecca Artursson
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Jakob Friedrich
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Max Backman
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Neda Hekmati
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Johanna Mattsson
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Cecilia Lindskog
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Johan Botling
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden; Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Artur Mezheyeuski
- Vall d'Hebron Institute of Oncology, Molecular Oncology Group, Barcelona, Spain; Vall d'Hebron Institute of Research, Barcelona, Spain
| | - Erika Broström
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Miklos Gulyas
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Klas Kärre
- Department of Microbiology, Cell and Tumor Biology, Karolinska Institutet, Stockholm, Sweden
| | - Johan Isaksson
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Carina Strell
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden; Centre of Cancer Biomarkers (CCBIO), Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
49
|
Liu M, Guan W, Xie X, Li Z, Qiu G, Lin X, Xie Z, Zhang J, Qin Y, Huang Z, Xu X, Zhou C. Phase I Clinical Trial of Autologous Hematopoietic Stem Cell Transplantation-Supported Dose-Intensified Chemotherapy With Adebrelimab as First-Line Treatment for Extensive-Stage Small Cell Lung Cancer. Clin Lung Cancer 2025; 26:e236-e241. [PMID: 39848827 DOI: 10.1016/j.cllc.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/26/2024] [Accepted: 12/26/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Small cell lung cancer (SCLC) is initially highly sensitive to chemotherapy, which often leads to significant tumor reduction. However, the majority of patients eventually develop resistance, and the disease is further complicated by its "cold" tumor microenvironment, characterized by low tumor immunogenicity and limited CD8+ T cell infiltration. These factors contribute to the poor response to immunotherapy in many cases of extensive-stage SCLC (ES-SCLC). High-dose chemotherapy has shown potential in enhancing tumor cytoreduction, but its use is often limited by severe hematologic toxicity. Combining chemotherapy with immune checkpoint inhibitors (ICIs) can create a synergistic effect by promoting immunogenic cell death and enhancing immune activation. Autologous hematopoietic stem cell transplantation (auto-HSCT) provides a means to support hematopoietic recovery, mitigate chemotherapy-induced myelosuppression, and contribute to immune reconstitution. In this context, the integration of auto-HSCT with dose-intensified chemotherapy and ICIs aims to both protect the bone marrow and enhance antitumor immune responses, potentially overcoming resistance to immunotherapy in ES-SCLC. METHODS A phase I, single-center, single-arm trial was designed to evaluate the safety and efficacy of auto-HSCT-supported dose-intensified chemotherapy combined with adebrelimab in treatment-naive ES-SCLC patients. Participants will receive induction chemotherapy followed by stem cell mobilization, apheresis, and cryopreservation. After successful mobilization, consolidation chemotherapy with stem cell reinfusion and granulocyte colony-stimulating factor (G-CSF) support will be performed. Maintenance therapy with adebrelimab continues until disease progression or unacceptable toxicity. Safety and efficacy data, including adverse events, objective response rate (ORR), progression-free survival (PFS), and overall survival (OS), will be analyzed. RESULTS The study aims to enhance treatment outcomes by overcoming resistance to immuno-chemotherapy and promoting immune reconstitution. The trial is ongoing at the First Affiliated Hospital of Guangzhou Medical University. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT06597513.
Collapse
Affiliation(s)
- Ming Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenhui Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaohong Xie
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zekun Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guihuan Qiu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinqing Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhanhong Xie
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiexia Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yinyin Qin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenqian Huang
- Department of Hematology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Xin Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Organ Transplantation, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Chengzhi Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
50
|
Moniwa K, Tokita S, Sumi T, Saijo H, Sugita S, Arioka K, Hirohashi Y, Chiba H, Kanaseki T, Torigoe T. Loss of Tapasin in Tumors Potentiates T-Cell Recognition and Anti-Tumor Effects of Immune Checkpoint Blockade. Cancer Sci 2025; 116:1203-1213. [PMID: 39989216 PMCID: PMC12044650 DOI: 10.1111/cas.70027] [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: 10/28/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 02/25/2025] Open
Abstract
Tumors can evade host immune surveillance by compromising the intracellular antigen processing machinery (APM), such as beta 2 macroglobulin (β2m) or the transporter associated with antigen processing (TAP). Defects in the APM generally result in the downregulation of surface MHC class I (MHC-I) levels. Here, we show that the downregulation of a component of the peptide loading complex (PLC), tapasin, in tumors conversely induces CD8+ T-cell responses and inhibits tumor growth in vivo. Loss of tapasin enhanced the anti-tumor effects of immune checkpoint blockade (ICB) in mouse non-small cell lung and colon cancer models. In contrast to β2m-deficient tumors, the reduced levels of MHC-I in tapasin-deficient tumors were restored by IFN-γ treatment, allowing them to be recognized by CD8+ T cells. These results suggest the presence of a reactive CD8+ T-cell fraction and the ability of immune surveillance to eliminate tumor variants with impaired tapasin expression.
Collapse
Affiliation(s)
- Keigo Moniwa
- Department of PathologySapporo Medical UniversitySapporoJapan
- Department of Respiratory Medicine and AllergologySapporo Medical UniversitySapporoJapan
| | - Serina Tokita
- Department of PathologySapporo Medical UniversitySapporoJapan
- Joint Research Center for ImmunoproteogenomicsSapporo Medical UniversitySapporoJapan
| | - Toshiyuki Sumi
- Department of Respiratory MedicineHakodate Goryoukaku HospitalHakodateJapan
| | - Hiroshi Saijo
- Department of Respiratory MedicineSapporo Minami‐Sanjo HospitalSapporoJapan
| | - Shintaro Sugita
- Department of Surgical PathologySapporo Medical UniversitySapporoJapan
| | - Kotomi Arioka
- Department of Surgical PathologyHakodate Goryoukaku HospitalHakodateJapan
| | | | - Hirofumi Chiba
- Department of Respiratory Medicine and AllergologySapporo Medical UniversitySapporoJapan
| | - Takayuki Kanaseki
- Department of PathologySapporo Medical UniversitySapporoJapan
- Joint Research Center for ImmunoproteogenomicsSapporo Medical UniversitySapporoJapan
| | | |
Collapse
|