101
|
Rolling CC, Lewirt S, Beitzen-Heineke A, Beckmann L, Bokemeyer C, Alsdorf W, Voigtlaender M, Langer F. Checkpoint Inhibitors, CAR T Cells, and the Hemostatic System: What Do We Know So Far? Hamostaseologie 2025; 45:175-187. [PMID: 40334710 DOI: 10.1055/a-2528-5071] [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: 05/09/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T cells are novel therapeutic strategies that enhance anticancer immunity by activating or engineering cancer-targeting T cells. The resulting hyperinflammation carries several side effects, ranging from autoimmune-like symptoms to cytokine release syndrome (CRS), with potentially severe consequences. Recent findings indicate that ICIs increase the risk of venous and arterial thromboembolic adverse events. Patients with prior VTE might be at higher risk of developing new events under ICI while other risk factors vary across studies. So far, data on CAR T-linked coagulopathies are limited. Hypofibrinogenemia in the presence of CRS is the most commonly observed dysregulation of hemostatic parameters. A rare but particularly severe adverse event is the development of disseminated intravascular coagulation activation, which can occur in the setting of CRS and may be linked to immune effector cell-associated hemophagocytic lymphohistiocytosis. While the increasing number of studies on thromboembolic complications and coagulation alterations under ICIs and CAR T therapies are concerning, these results might be influenced by the retrospective study design and the heterogeneous patient populations. Importantly, numerous promising new T cell-based immunotherapies are currently under investigation for various cancers and are expected to become very prominent therapy options in the near future. Therefore, coagulopathies and thrombosis under T cell-directed immuno- and anti-cancer therapies is important. Our review provides an overview of the current understanding of ICI- and CAR T-associated thromboembolism. We discuss pathogenic mechanisms of inflammation-associated coagulation activation and explore potential biomarkers for VTE.
Collapse
Affiliation(s)
- Christina C Rolling
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Lewirt
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Beitzen-Heineke
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, United States
| | - Lennart Beckmann
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Winfried Alsdorf
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Minna Voigtlaender
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Langer
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
102
|
Rausch L, Kallies A. Molecular Mechanisms Governing CD8 T Cell Differentiation and Checkpoint Inhibitor Response in Cancer. Annu Rev Immunol 2025; 43:515-543. [PMID: 40279308 DOI: 10.1146/annurev-immunol-082223-044122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
CD8 T cells play a critical role in antitumor immunity. However, over time, they often become dysfunctional or exhausted and ultimately fail to control tumor growth. To effectively harness CD8 T cells for cancer immunotherapy, a detailed understanding of the mechanisms that govern their differentiation and function is crucial. This review summarizes our current knowledge of the molecular pathways that regulate CD8 T cell heterogeneity and function in chronic infection and cancer and outlines how T cells respond to therapeutic checkpoint blockade. We explore how T cell-intrinsic and -extrinsic factors influence CD8 T cell differentiation, fate choices, and functional states and ultimately dictate their response to therapy. Identifying cells that orchestrate long-term antitumor immunity and understanding the mechanisms that govern their development and persistence are critical steps toward improving cancer immunotherapy.
Collapse
Affiliation(s)
- Lisa Rausch
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia;
| | - Axel Kallies
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia;
| |
Collapse
|
103
|
Gray R, Manisty C, Cheng R, Dastidar A, Mamas M, Ghosh A. Immune checkpoint inhibitors: Unravelling atherosclerotic cardiovascular risk. Atherosclerosis 2025; 403:119147. [PMID: 40037087 DOI: 10.1016/j.atherosclerosis.2025.119147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/02/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Affiliation(s)
| | | | - Richard Cheng
- Division of Cardiology, University of Washington, USA
| | - Amardeep Dastidar
- Bristol Heart Institute, NIHR Biomedical Research Centre Cardiovascular & North Bristol NHS Trust, Bristol, UK
| | - Mamas Mamas
- Keele Cardiovascular Research Group, Keele University, UK
| | | |
Collapse
|
104
|
Dong Y, Li J, Dai Y, Zhang X, Wang T, Zhao B, Liu W, Chen L, Yang S, Du P, Jiao Z. Redox-responsive metal-organic framework nanocapsules enhance tumor chemo-immunotherapy by modulating tumor metabolic reprogramming. Mater Today Bio 2025; 31:101487. [PMID: 39896279 PMCID: PMC11786678 DOI: 10.1016/j.mtbio.2025.101487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/04/2025] [Accepted: 01/11/2025] [Indexed: 02/04/2025] Open
Abstract
Immunotherapy, particularly immune checkpoint blockade (ICB), has opened the era of modern oncology, offering significant promise for modern oncology. However, the efficacy of immunotherapy is frequently curtailed by the immunosuppressive tumor microenvironment (ITM), a milieu shaped by tumor metabolic reprogramming. Herein, a novel tumor microenvironment-responsive nanocapsules (DNMCs) were developed that simultaneously modulate tumor metabolism and the ITM to enhance the effectiveness of chemo-immunotherapy. DNMCs consist of an acidic and redox-sensitive metal-organic framework (MOF) encapsulating Doxorubicin (DOX) and the indoleamine-2,3-dioxygenase1 (IDO1) inhibitor NLG919. In the tumor microenvironment, DNMCs degrade, rapidly releasing DOX and NLG919. DOX induces immunogenic cell death (ICD), while NLG919 regulates amino acid metabolism by modulating IDO1 activity, thereby reversing the immunosuppressive of ITM. Consequently, DNMCs elicit effective anti-tumor immune responses, characterized by an increased density of tumor-infiltrating CD8+ cytotoxic T cells as well as depletion of immunosuppressive regulatory T cells (Tregs), thus effectively suppressing pancreatic cancer growth in KPC mice through combined chemo-immunotherapy. Overall, DNMCs exhibit significant tumor growth inhibition in pancreatic cancer patient-derived organoids (PDOs) and mouse models. This study presents a promising approach to enhancing chemo-immunotherapy by targeting tumor metabolic reprogramming and augmenting immune response against malignant tumors.
Collapse
Affiliation(s)
- Yuman Dong
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Jieru Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Yiwei Dai
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Xinyu Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Tao Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Bin Zhao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Wenbo Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, China
| | - Li Chen
- Department of Orthopaedics, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Shaopei Yang
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Pengcheng Du
- College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Zuoyi Jiao
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| |
Collapse
|
105
|
Zhang T, Tang D, Wu P, Jiang S, Zhang Y, Naeem A, Li Y, Li C, Hu B, Guo S, Sun C, Xiao H, Yan R, Weng Y, Huang Y. NIR-II photo-accelerated polymer nanoparticles boost tumor immunotherapy via PD-L1 silencing and immunogenic cell death. Bioact Mater 2025; 46:285-300. [PMID: 39811466 PMCID: PMC11732249 DOI: 10.1016/j.bioactmat.2024.12.018] [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: 08/12/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Immune checkpoint blockade (ICB) therapy is a widely favored anti-tumor treatment, but it shows limited response to non-immunogenic "cold" tumors and suffers from drug resistance. Photodynamic therapy (PDT), as a powerful localized treatment approach, can convert a "cold tumor" into a "hot tumor" by inducing immunogenic cell death (ICD) in tumor cells, thereby enhancing tumor immunogenicity and promoting tumor immunotherapy. However, the effectiveness of PDT is largely hindered by the limited penetration depth into tumor tissues. To address these issues, we proposed an all-in-one drug system with NIR-II photo-accelerated PDT effects, efficient immune checkpoint gene silencing, and a facile manufacturing process. The so-called all-in-one drug system comprises a multi-modal designed polymer PPNP and siRNA. PPNP is an amphipathic polymer that includes the near infrared-II (NIR-II) photosensitizer Aza-boron-dipyrromethene (Aza-BODIPY), a glutathione (GSH)-cleavable linker, and a cationic monomer derived from cholesterol. PPNP can self-assemble and efficiently load siRNA. Under laser irradiation, PPNP triggers a potent ICD cascade, causing the on-demand release of siPD-L1, reshaping the tumor's immunosuppressive microenvironment, effectively inhibiting the growth of various tumors, and stimulating the immune memory. This study represents a generalized platform for PDT and gene silencing, designed to modulate immune-related signaling pathways for improved anticancer therapy.
Collapse
Affiliation(s)
- Tian Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Science Laboratory of Polymer Physics and Chemistry Institute of Chemistry Chinese Academy of Science Beijing 100190, China
| | - Pengfei Wu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Shaoping Jiang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuquan Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Abid Naeem
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Yong Li
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Chunhui Li
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Bo Hu
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Shuai Guo
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Caixia Sun
- School of Chemistry, Chemical Engineering & Biotechnology, Nanyang Technological University, 637371, Singapore
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Science Laboratory of Polymer Physics and Chemistry Institute of Chemistry Chinese Academy of Science Beijing 100190, China
| | - Ran Yan
- Key Laboratory of Biomedical Functional Materials, School of Science, China Pharmaceutical University, Nanjing, 211198, China
| | - Yuhua Weng
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology (BIT), Zhuhai 519088, China
- Advanced Technology Research Institute, Beijing Institute of Technology (BIT), Jinan 250101, China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology (BIT), Zhuhai 519088, China
- Advanced Technology Research Institute, Beijing Institute of Technology (BIT), Jinan 250101, China
| |
Collapse
|
106
|
Yang Y, Xu Q, Gu W, Nan K, Chen S, Wang S, Zhang J, Zhao Q. Oxidative stress-augmented Cu-doped hollow mesoporous carbon nanozyme for photothermal/photodynamic synergistic therapy. J Colloid Interface Sci 2025; 683:910-925. [PMID: 39709766 DOI: 10.1016/j.jcis.2024.12.076] [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: 09/12/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Photodynamic therapy (PDT) has witnessed remarkable progress in recent years owing to its specific properties. Given that the antioxidation system of tumor microenvironment (TME) adversely affects treatment outcomes, powerful TME modulators can significantly resolve the limitation of PDT. Herein, we developed a PEG-modified Cu2+-doped hollow mesoporous carbon nanozyme (CHC-PEG) and loaded insoluble photosensitizer IR780 into its pores and cavities to construct the multifunctional nano-system IR780/CHCP. CHC-PEG nanozyme could perform photothermal therapy (PTT) effect and protect IR780 from aggregation-caused quenching (ACQ) effect, while exerting peroxidase (POD)-mimetic activity and the ability of consuming glutathione (GSH) to achieve oxidative stress-augmented PDT effect. When exposed to near-infrared (NIR) light, IR780 was stimulated to produce singlet oxygen (1O2) and CHC-PEG could increase the temperature of TME to exert stronger POD-mimetic activity for producing more hydroxyl radicals (OH), therefore the IR780/CHCP nano-system exhibited remarkable tumor growth inhibition. Benefited by the enhanced synergistic effect, IR780/CHCP exhibited remarkable in vivo tumor growth inhibition, with the tumor inhibition rate of 93 %, and had no significant effect on major organs. Above all, IR780/CHCP could resist the antioxidant system in TME to enhance the level of oxidative stress, thereby enabling effective anti-tumor therapy. This study introduced a novel strategy to effectively promote the synergistic PTT/PDT effect by the enhanced oxidative stress.
Collapse
Affiliation(s)
- Yuanqi Yang
- Department of Microbial and Biochemical Pharmacy, School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Qingqing Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Wei Gu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Kaisheng Nan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Siyu Chen
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China
| | - Jinghai Zhang
- Department of Microbial and Biochemical Pharmacy, School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning Province 110016, PR China.
| |
Collapse
|
107
|
Mouri A, Imai H, Endo S, Nakagawa J, Tsukamoto K, Kurata Y, Yamaguchi O, Masaki K, Hashimoto K, Shiono A, Miura Y, Kobayashi K, Kaira K, Kagamu H. A Real-Word Analysis of the Correlation Between Clinical Efficacy and Predictive Factors of Immune-Related Adverse Events in Patients With Nonsmall Lung Cancer Treated With Nivolumab Plus Ipilimumab. Cancer Med 2025; 14:e70741. [PMID: 40249663 PMCID: PMC12007460 DOI: 10.1002/cam4.70741] [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: 09/30/2024] [Revised: 02/15/2025] [Accepted: 02/25/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND The combination of nivolumab and ipilimumab, which act on different immune checkpoint molecules, is a promising first-line treatment strategy for advanced nonsmall cell lung cancer (NSCLC). However, real-world clinical data on this regimen, particularly regarding the relationship between adverse events (AEs) and efficacy, are inadequate. METHODS This real-world retrospective study was conducted on patients with advanced or recurrent NSCLC treated using a combination of nivolumab and ipilimumab as a first-line treatment. We extracted the data of consecutive eligible patients from four institutions in Japan between December 2020 and November 2022. RESULTS The study population comprised 184 patients who received nivolumab plus ipilimumab (median follow up period: 13.0 months [0.3-35.0]). In total, 81.0% (n = 149) of the patients were men, and the median age was 72.0 years (range: 46-80). The median progression-free survival (PFS) and overall survival (OS) were 6.6 months (95% confidence interval [CI]: 4.7-8.2) and 17.4 months (95% CI: 11.9-20.4), respectively. Skin disorders, liver dysfunction, thyroid dysfunction, and pneumonitis were the most common adverse events (AEs), with AEs occurring in 154 patients (83.7%). The median PFS in the AE group was longer than that in the non-AE group (8.2 vs. 2.6 months, p < 0.0001). The median OS in the AE group was also better than that in the non-AE group (19.3 vs. 6.1 months, p < 0.0001). Multivariate logistic regression analysis identified smoking history and high PD-L1 expression as factors related to the incidence of grade 3 and 4 AEs, respectively. The incidence of multiple AEs revealed a significant association with a longer PFS and OS. Skin disorders, adrenal insufficiency, and eosinophilia were the AEs with the greatest impact on survival. CONCLUSIONS Patients who experienced AEs had significantly longer PFS. Among AEs, the occurrence of skin disorders, adrenal insufficiency, and eosinophilia were likely to prolong PFS and OS.
Collapse
Affiliation(s)
- Atsuto Mouri
- Department of Respiratory Medicine, International Medical CenterSaitama Medical UniversityHidakaJapan
| | - Hisao Imai
- Department of Respiratory Medicine, International Medical CenterSaitama Medical UniversityHidakaJapan
| | - Satoshi Endo
- Division of Respiratory MedicineGunma Prefectural Cancer CenterOtaJapan
| | - Junichi Nakagawa
- Department of Respiratory MedicineNational Hospital Organization Takasaki General Medical CenterTakasakiJapan
| | - Kasumi Tsukamoto
- Department of PulmonologyNational Hospital Organization Disaster Medical CenterTokyoJapan
| | - Yuhei Kurata
- Department of Respiratory Medicine, International Medical CenterSaitama Medical UniversityHidakaJapan
- Division of Infectious Diseases and Respiratory Medicine, Department of Internal MedicineNational Defense Medical CollegeTokorozawaJapan
| | - Ou Yamaguchi
- Department of Respiratory Medicine, International Medical CenterSaitama Medical UniversityHidakaJapan
| | - Kenji Masaki
- Department of Respiratory Medicine, International Medical CenterSaitama Medical UniversityHidakaJapan
| | - Kosuke Hashimoto
- Department of Respiratory Medicine, International Medical CenterSaitama Medical UniversityHidakaJapan
| | - Ayako Shiono
- Department of Respiratory Medicine, International Medical CenterSaitama Medical UniversityHidakaJapan
| | - Yu Miura
- Department of Respiratory Medicine, International Medical CenterSaitama Medical UniversityHidakaJapan
| | - Kunihiko Kobayashi
- Department of Respiratory Medicine, International Medical CenterSaitama Medical UniversityHidakaJapan
| | - Kyoichi Kaira
- Department of Respiratory Medicine, International Medical CenterSaitama Medical UniversityHidakaJapan
| | - Hiroshi Kagamu
- Department of Respiratory Medicine, International Medical CenterSaitama Medical UniversityHidakaJapan
| |
Collapse
|
108
|
Ye H, Li M. Baseline (modified) Glasgow prognostic score as a predictor of therapeutic response to immune checkpoint inhibitors in solid tumors: A systematic review and meta‑analysis. Oncol Lett 2025; 29:184. [PMID: 40007624 PMCID: PMC11851447 DOI: 10.3892/ol.2025.14931] [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: 10/15/2024] [Accepted: 01/15/2025] [Indexed: 02/27/2025] Open
Abstract
A systemic analysis was performed to evaluate the prognostic utility of the Glasgow prognostic score (GPS) and the modified (m)GPS in cancer patients treated with immune checkpoint inhibitors (ICI). The PubMed, Cochrane Library, EMBASE and Google Scholar databases were searched for entries added until May 1st, 2023, to obtain relevant articles for this study. The analysis examined several clinical outcomes, including overall survival (OS), progression-free survival (PFS), objective response rate and disease control rate (DCR). In this analysis, a total of 38 articles with 3,772 patients were included. The pooled results indicated that patients with high GPS levels had shorter OS [GPS 2 vs. 0, hazard ratio (HR): 4.35, P<0.001; GPS 1 vs. 0, HR: 2.00, P<0.001; GPS 2 vs. 1/0, HR: 2.62, P<0.001; GPS 2/1 vs. 0, HR: 2.60, P<0.001) and PFS (GPS 2 vs. 0, HR: 2.11, P=0.001; GPS 1 vs. 0, HR: 1.33, P=0.001; GPS 2 vs. 1/0, HR: 2.11, P<0.001; GPS 2/1 vs. 0, HR: 1.62, P<0.001], as well as a lower DCR [GPS 2 vs. 1/0, odds ratio (OR): 0.53, P<0.001, GPS 2/1 vs. 0, OR: 0.51, P<0.001]. It was also found that patients with high mGPS levels had poorer OS (mGPS 2 vs. 0, HR: 3.15, P<0.001; mGPS 1 vs. 0, HR: 1.70, P<0.001; mGPS 2 vs. 1/0, HR: 1.95, P=0.049; mGPS 2/1 vs. 0, HR: 3.14, P=0.041; continuous variables, HR: 1.52, P<0.001) and PFS (mGPS 2 vs. 0, HR: 2.70, P<0.001; mGPS 1 vs. 0, HR: 1.74, P=0.016; mGPS 2 vs. 1/0, HR: 1.91, P=0.044; continuous variables, HR: 1.29, P<0.001), and lower DCR (mGPS 2 vs. 1/0, HR: 0.46, P<0.001). In conclusion, the GPS and mGPS were reliable predictors of outcomes in cancer patients treated with ICIs.
Collapse
Affiliation(s)
- Hong Ye
- Department of Respiratory and Critical Care Medicine, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Meifang Li
- Department of Brain Disease, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
109
|
Torrisi R, Gerosa R, Miggiano C, Saltalamacchia G, Benvenuti C, Santoro A. Beyond failure of endocrine-based therapies in HR+/HER2 negative advanced breast cancer: What before chemotherapy? A glimpse into the future. Crit Rev Oncol Hematol 2025; 208:104634. [PMID: 39900320 DOI: 10.1016/j.critrevonc.2025.104634] [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: 11/04/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Despite the impressive improvements achieved by endocrine therapy and CDK4/6 inhibitors (CDK4/6i) and the forthcoming availability of alternative endocrine manipulations and targeted therapies, hormone-receptor positive/HER2 negative (HR+/HER2-) advanced breast cancer (ABC) is almost inevitably destined to become endocrine- refractory. At this time chemotherapy has been recently challenged and partly replaced by new targeted options as antibody-drug conjugated (ADCs). Trastuzumab-deruxtecan has been proven meaningfully superior to chemotherapy either in 1st and later lines after progression to CDK4/6i in HER2-low ABC and results with other ADCs as Sacituzumab Govitecan and Datopotamab-deruxtecan are promising, but the definition of cross-resistance between these drugs sharing either antibody or payload is crucial before implementing them in a useful sequence. While PARP inhibitors are the standard 2nd line in patients with gBRCA mutation, it is not still known whether patients with mutations of PALB2 or of other homologous recombinant defect (HRD)-related genes will benefit of the same treatment. On the other hand, the results obtained with immune checkpoint inhibitors (ICIs) in HR+ /HER2-ABC contrarily to the early setting are disappointing up to now, but investigations of ICIs in combination with other targeted drugs which may increase immune response and the search for better markers of activity are under way. Moreover the anticipation in upfront treatment of ADCs or PARPi in patients with features of putative endocrine resistance and/or of less sensitiviy to CDK4/6i and the choice of therapy in patients recurring during or soon after adjuvant CDK4/6i and olaparib represent further challenges for the future.
Collapse
Affiliation(s)
- Rosalba Torrisi
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy.
| | - Riccardo Gerosa
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Chiara Miggiano
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Giuseppe Saltalamacchia
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy
| | - Chiara Benvenuti
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| | - Armando Santoro
- Humanitas Research Hospital IRCCS, Medical Oncology and Hematology Unit, Viale Manzoni 56, Rozzano, MI 20089, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
| |
Collapse
|
110
|
Adhikari A, Chen IA. Antibody-Nanoparticle Conjugates in Therapy: Combining the Best of Two Worlds. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409635. [PMID: 40051146 PMCID: PMC12001320 DOI: 10.1002/smll.202409635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/02/2025] [Indexed: 04/17/2025]
Abstract
Monoclonal antibodies (mAbs) and antibody fragments have revolutionized medicine as highly specific binding agents and inhibitors. At the same time, several types of nanomaterials, including liposomes, lipid nanoparticles (NPs), polymersomes, metal and metal oxide NPs, and protein nanostructures, are increasingly utilized and explored for therapeutic potential due to their versatility, chemical and physical properties, and tunability. However, nanomaterials alone often lack specificity, leading to relatively low efficacy and/or high toxicity. To address this problem, a rapidly emerging area is antibody-nanomaterial conjugates (ANCs), which combine the precise targeting specificity of antibodies with the effector functionality of the nanomaterial. In this review, we give a brief introduction to mAbs and major conjugation techniques, describe major classes of nanomaterials being studied for therapeutic potential, and review the literature on ANCs of each class. Special focus is given to emerging applications including ANCs addressing the blood-brain barrier, ANCs delivering nucleic acids, and light-activated ANCs. While many disease targets are related to cancer, ANCs are also under development to address autoimmune, neurological, and infectious diseases. While important challenges remain, ANCs are poised to become a next-generation therapeutic technology.
Collapse
Affiliation(s)
- Aniruddha Adhikari
- Department of Chemical and Biomolecular EngineeringDepartment of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCA90049USA
| | - Irene A. Chen
- Department of Chemical and Biomolecular EngineeringDepartment of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCA90049USA
| |
Collapse
|
111
|
Tang FH, Wong HYT, Tsang PSW, Yau M, Tam SY, Law L, Yau K, Wong J, Farah FHM, Wong J. Recent advancements in lung cancer research: a narrative review. Transl Lung Cancer Res 2025; 14:975-990. [PMID: 40248731 PMCID: PMC12000946 DOI: 10.21037/tlcr-24-979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/27/2025] [Indexed: 04/19/2025]
Abstract
Background and Objective Lung cancer remains the leading cause of cancer-related mortality worldwide, with a 5-year survival rate ranging from 10% to 20%. The majority of cases are categorized as non-small cell lung cancer (NSCLC) (80%) and small cell lung cancer (SCLC) (20%), with NSCLC being the more prevalent type. Tobacco use, particularly cigarette smoking, is a significant contributor to over 80% of lung cancer cases. Early diagnosis is challenging due to limitations in screening methods, resulting in many cases being identified only in advanced stages. Moreover, current treatment options often exhibit low efficacy, partly due to an inadequate understanding of the disease's pathogenesis. This narrative review aims to summarize recent discoveries and advancements in lung cancer research, focusing on improvements in diagnosis, treatment, and understanding of the disease. Methods A comprehensive literature review was performed utilizing the PubMed Central database to identify recent studies relevant to lung cancer. This review synthesizes findings from various research articles to provide a cohesive summary of advancements in the field. Key Content and Findings In the past decade, notable progress has been achieved in lung cancer research, particularly concerning diagnostics and treatment strategies. Novel therapeutic approaches, including immunotherapy and genomic-targeted therapies, have demonstrated promising results. Understanding the tumor microenvironment (TME) and the role of T lymphocytes has become crucial for developing effective treatments. Additionally, advancements in immune checkpoint inhibitors (ICIs) have shown potential in enhancing patient outcomes. Improvements in tumor detection technologies are also anticipated to facilitate earlier diagnosis, ultimately contributing to better survival rates. Conclusions Significant strides have been made in lung cancer research over the last ten years, particularly in diagnostics and treatment methodologies. Future research should prioritize exploring the TME, the function of T lymphocytes, and the efficacy of ICIs while continuing to innovate in tumor detection technologies. Such efforts are essential for enhancing treatment outcomes and improving the overall quality of life for lung cancer patients.
Collapse
Affiliation(s)
- Fuk Hay Tang
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| | - Heylie Y. T. Wong
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| | | | - Mabel Yau
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| | - Shing Yau Tam
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| | - Lawla Law
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| | - Katherine Yau
- School of Nursing, Tung Wah College, Hong Kong, China
| | - Jade Wong
- Library, Tung Wah College, Hong Kong, China
| | | | - Jacky Wong
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China
| |
Collapse
|
112
|
Fang Z, Liao SC, Guo YY, Li JJ, Wang Z, Zhang YM, Yao F. Development of coenzyme Q10-related molecular subtypes and a prognostic signature for predicting breast cancer prognosis and response to immunotherapy. Transl Cancer Res 2025; 14:2010-2028. [PMID: 40225007 PMCID: PMC11985207 DOI: 10.21037/tcr-2025-425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025]
Abstract
Background Breast cancer (BRCA) remains by far the most life-threatening malignancy in women. Resistance to BRCA treatment may be counteracted by the induction of ferroptosis in combination with immunotherapy. The study aims develop iron death-related prognostic models to predict prognosis and immunotherapy effects in BRCA patients. Methods We collected and organized 22 ferroptosis-related pathways and quantified their pathway activities using single-sample gene set enrichment analysis (ssGSEA). Coenzyme Q10 (CoQ10) is a pathway associated with prognosis in patients with BRCA. We compared the differences between patients with different CoQ10 expressions in terms of prognosis, biological function, mutational profile, immune infiltration, immunotherapy, and chemotherapeutic drug sensitivity. Results Patients with high CoQ pathway activity had a worse prognosis. In addition, patients with high CoQ activity showed greater cell cycle activation and lower immune infiltration. Based on different CoQ10 expression patterns, we developed a CoQ10-related prognostic model. The accuracy and stability of CoQ10-related prognostic models were well validated in the training set and multiple validation sets. High-risk patients showed a propensity for immune depletion and tolerance to immunotherapy. There were also some differences in the sensitivity to different chemotherapeutic agents between high- and low-risk patients. Conclusions We have constructed and validated a CoQ10-related gene model that can predict the prognosis of BRCA. Critically, it may serve as a reference standard to guide outcome prognostication in patients with BRCA.
Collapse
Affiliation(s)
- Zhou Fang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shi-Chong Liao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yue-Yue Guo
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Juan-Juan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong Wang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi-Min Zhang
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Feng Yao
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
113
|
Tong L, Yuan Y, He W, Yang W, Pan X. Adverse events associated with acute pancreatitis caused by immune checkpoint inhibitors: a pharmacovigilance analysis of the FDA adverse event reporting system (FAERS) database. Expert Opin Drug Saf 2025:1-9. [PMID: 40152025 DOI: 10.1080/14740338.2025.2486311] [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: 08/25/2024] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND The precise incidence of immune-related adverse events (irAEs) remains unclear. This pharmacovigilance study investigated acute pancreatitis (AP) associated with immune checkpoint inhibitors (ICIs) using real-world data from the FDA Adverse Event Reporting System (FAERS). RESEARCH DESIGN AND METHODS Disproportionality analysis employing reporting odds ratios (RORs) was conducted to detect AP signals in ICI-treated patients compared to the entire FAERS database. RESULTS A total of 152,042 individual patients were included in the dataset from which we identified a cohort of 921 acute pancreatitis adverse events (AEs). The severe outcome of acute pancreatitis was death, with a rate of 13.6% (125/921). Immune checkpoint inhibitor (ICI)-related acute pancreatitis AEs were classified into two categories (pancreatitis and immune-mediated pancreatitis) based on the type of adverse event observed. ICI treatments were significantly correlated with the risk of ICIs-induced acute pancreatitis (AP) but varied among different drugs. The median time to AP onset was 57 days, with events occurring throughout the first year post-ICI initiation. CONCLUSIONS Our findings provide an enhanced understanding of potential acute pancreatitis related adverse events and provide actionable insights for the early detection and management of ICI related pancreatic adverse events.
Collapse
Affiliation(s)
- Lihua Tong
- Department of Oncology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Yanling Yuan
- Department of Oncology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Wanming He
- Department of Oncology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Wen Yang
- Department of Oncology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| | - Xingxi Pan
- Department of Oncology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangdong, China
| |
Collapse
|
114
|
Zhao S, Kang S. Comprehensive economic value evaluation of adding tislelizumab to first-line chemotherapy for patients with extensive-stage small cell lung cancer in China. Expert Rev Pharmacoecon Outcomes Res 2025:1-7. [PMID: 40134070 DOI: 10.1080/14737167.2025.2483423] [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: 11/10/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND The cost-effectiveness of tislelizumab plus chemotherapy for patients with extensive-stage small cell lung cancer (ES-SCLC) was uncleared yet. The current analysis aimed to evaluate the cost-effective of adding tislelizumab to the first-line chemotherapy for patients with untreated ES-SCLC from the Chinese health-care system perspective. RESEARCH DESIGN AND METHODS A partitioned survival model that simulated 3-week patients transition in 10-year time horizon was established to evaluate the economic value. The clinical benefit and safety data were gathered from the RATIONALE-312 trial, model parameters were gathered from the local charges and previously published studies. Sensitivity analyses and subgroup analyses were conducted to examine the robustness of the model outcomes. RESULTS Tislelizumab plus chemotherapy could bring additional 0.61 LYs and 0.30 QALYs with the marginal cost of $5,849.40, resulting in the incremental cost-effectiveness ratio (ICER) of $19,592.08 per additional quality-adjusted life-years (QALYs) gained. Sensitivity analyses and subgroup analyses confirmed the robustness of the model results for both intention-to-treat patients and all subgroup patients. CONCLUSIONS Tislelizumab plus chemotherapy could be considered cost-effective first-line therapy for patients with untreated ES-SCLC patients compared with chemotherapy alone from the perspective of Chinese health-care system.
Collapse
Affiliation(s)
- Shan Zhao
- Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China
| | - Shuo Kang
- Medical Insurance Office, The Second Hospital of Hebei Medical University, Shijiazhuang, PR China
| |
Collapse
|
115
|
Pereyra Pietri M, Farina JM, Scalia IG, Mahmoud AK, Roarke M, Wasef B, Tagle-Cornell C, Kenyon CR, Abbas MT, Ali NB, Awad KA, Javadi N, Bismee NN, Larsen CM, Herrmann J, Arsanjani R, Ayoub C. Comparison of the diagnostic and prognostic value of criteria for immune checkpoint inhibitor related myocarditis. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:30. [PMID: 40149000 PMCID: PMC11948924 DOI: 10.1186/s40959-025-00327-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Myocarditis is a dreaded complication of immune-checkpoint inhibitor (ICI) therapy but challenging to diagnose. There are no published data comparing the two leading diagnostic criteria for ICI-related myocarditis (ICIrM) and their association with cardiovascular events. METHODS In this retrospective cohort study, we reviewed all patients who underwent ICI therapy and had cardiac troponin assessment for possible myocarditis across three tertiary institutions from 2011 to 2022. ICIrM was adjudicated by the Bonaca et al. criteria and the ESC-ICOS guidelines. A propensity matched control group was identified of patients treated with ICI without developing myocarditis. Baseline characteristics and long-term outcomes, including cardiac death, MACE (myocardial infarction, TIA/stroke, heart failure), and arrhythmias data were curated, and patients diagnosed with ICIrM by each criteria were compared to controls for cardiovascular events. RESULTS A total of 59 patients (mean age was 73.1 ± 10.2 years, 60.1% male) were identified as having a diagnosis of ICIrM by Bonaca criteria (16 definite, 13 probable and 30 possible myocarditis). Forty-seven of these patients met the ESC-ICOS guidelines criteria, and all patients meeting either set of ICIrM criteria were treated with steroid therapy. At 3-year follow up, patients diagnosed with ICIrM by the Bonaca criteria had a high risk of cardiac mortality (HR 17.84, 95%CI 2.36-134.62, p = 0.005), MACE (HR 4.90, 95%CI 2.40-10.02, p < 0.001) and arrhythmias (HR 3.33, 95%CI 1.78-6.21, p < 0.001) when compared to matched controls. ICIrM by ESC-ICOS criteria was similarly predictive of cardiac mortality, MACE, and arrhythmias (HR 15.01, 95%CI 1.96-114.76, p = 0.009, HR 5.18, 95%CI 2.33-11.53, p < 0.001, and HR 3.41, 95%CI 1.73-6.70, p < 0.001 respectively). CONCLUSION The ESC-ICOS guidelines were more restrictive than the Bonaca et al. criteria for the diagnosis of ICIrM but similar in terms of prognostic value.
Collapse
Affiliation(s)
- Milagros Pereyra Pietri
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Juan M Farina
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Isabel G Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Ahmed K Mahmoud
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Michael Roarke
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Beman Wasef
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Cecilia Tagle-Cornell
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Courtney R Kenyon
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Mohammed Tiseer Abbas
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Nima Baba Ali
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Kamal A Awad
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Niloofar Javadi
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Nadera N Bismee
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Carolyn M Larsen
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA.
| |
Collapse
|
116
|
Lovewell RR, Langermann S, Flies DB. Immune inhibitory receptor agonist therapeutics. Front Immunol 2025; 16:1566869. [PMID: 40207220 PMCID: PMC11979287 DOI: 10.3389/fimmu.2025.1566869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
The immune system maintains the health of an organism through complex sensing and communication mechanisms. Receptors on the surface of immune cells respond to stimuli resulting in activity described at its most basic as inhibitory or stimulatory. Significant progress in therapeutic intervention has occurred by modulating these pathways, yet much remains to be accomplished. Therapeutics that antagonize, or block, immune inhibitory receptor (IIR) pathways, such as checkpoint inhibitors in cancer are a key example. Antagonism of immune stimulatory receptors (ISRs) for dysregulated inflammation and autoimmunity have received significant attention. An alternative strategy is to agonize, or induce signaling, in immune pathways to treat disease. Agonism of ISRs has been employed with some success in disease settings, but agonist therapeutics of IIRs have great, untapped potential. This review discusses and highlights recent advances in pre-clinical and clinical therapeutics designed to agonize IIR pathways to treat diseases. In addition, an understanding of IIR agonists based on activity at a cellular level as either agonist suppression of stimulatory cells (SuSt), or a new concept, agonist suppression of suppressive cells (SuSu) is proposed.
Collapse
|
117
|
Xu F, Sang J, Wang N, Wang M, Huang Y, Ma J, Chen H, Xie Q, Wei Z, Ye X. Microwave ablation combined with immune checkpoint inhibitor enhanced the antitumor immune activation and memory in rechallenged tumor mouse model. Cancer Immunol Immunother 2025; 74:161. [PMID: 40131498 PMCID: PMC11937475 DOI: 10.1007/s00262-025-04003-5] [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: 12/29/2024] [Accepted: 02/26/2025] [Indexed: 03/27/2025]
Abstract
Microwave ablation (MWA) is a super minimally invasive therapeutic approach that has been widely applied in the treatment of non-small cell lung cancer (NSCLC). Although MWA can elicit antitumor immune responses, these immune responses are not relatively steady and insufficient to completely clear recurrence tumor cells within the body. Immunotherapy monotherapy has shown low clinical efficacy in the treatment of advanced NSCLC. MWA combined with immune checkpoint inhibitors (ICIs) is a promising therapeutic approach. However, the mechanism of synergic effect remains elusive. In this study, we have conducted a retrospective analysis of the clinical outcomes of MWA combined with ICIs, finding that the combinational therapy yielded superior Objective Response Rate and longer Progression-Free Survival. In preclinical models, we established a tumor rechallenged model to address post-MWA recurrence and to delve into the underlying mechanisms of the combined therapy. We observed that the combined treatment (MWA + PD-L1 blockade therapy) effectively addressed the issue of tumor recurrence in tumor rechallenged model. The combinational therapy increased the function and percentage of CD8+ tumor-infiltrating lymphocytes, enhanced the functionality of CD8+ T cells within tumor-draining lymph nodes (TdLNs), and elevated the proportion of T central memory cells. Additionally, the combined treatments promoted the proportion of Migration Dendritic Cells type 1 (Mig DC1) within TdLNs, thereby enhancing their activation potential. Notably, FTY720-mediated blockade of lymphocyte egress abolished the therapeutic benefits, confirming TdLNs-dependent systemic immunity. Moreover, the efficacy of the combinational therapy depended on the migration of T cells from TdLNs to tumor site. In summary, we proposed a potentially effective combined treatment regimen and have elucidated the underlying cellular mechanisms that underpin its efficacy.
Collapse
Affiliation(s)
- Fengkuo Xu
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Jing Sang
- Department of Pathology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, 271000, Shandong, China
| | - Nan Wang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Shandong Provincial Qianfoshan Hospital, Jinan, 250014, Shandong, China
| | - Meixiang Wang
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | | | - Ji Ma
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
| | - Huanan Chen
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China
- Department of Health Management, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong. Engineering Laboratory for Health Management, Shandong Medicine and Health Key Laboratory of Laboratory Medicine, Jinan, 250014, China
- Department of Medical Record Management and Statistics, The First Affiliated Hospital of Shandong First Medical University &Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Qi Xie
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
- Shandong Provincial Lab for Clinical Immunology Translational Medicine in Universities, Jinan, 250014, China.
| | - Zhigang Wei
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
- Cheeloo College of Medicine, Shandong University, Jinan, 250033, China.
| | - Xin Ye
- Department of Oncology, Lung Cancer Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, 250014, China.
- Shandong Provincial Lab for Clinical Immunology Translational Medicine in Universities, Jinan, 250014, China.
| |
Collapse
|
118
|
Berry D, Moldoveanu D, Rajkumar S, Lajoie M, Lin T, Tchelougou D, Sakthivel S, Sharon I, Bernard A, Pelletier S, Ripstein Y, Spatz A, Miller WH, Jamal R, Lapointe R, Mes-Masson AM, Petrecca K, Meguerditchian AN, Richardson K, Wang B, Chergui M, Guiot MC, Watters K, Stagg J, Schmeing TM, Rodier F, Turcotte S, Mihalcioiu C, Meterissian S, Watson IR. The NF1 tumor suppressor regulates PD-L1 and immune evasion in melanoma. Cell Rep 2025; 44:115365. [PMID: 40023845 DOI: 10.1016/j.celrep.2025.115365] [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: 10/20/2023] [Revised: 11/24/2024] [Accepted: 02/06/2025] [Indexed: 03/04/2025] Open
Abstract
Hotspot BRAF, hotspot NRAS, and NF1 loss-of-function mutations are found in approximately 50%, 25%, and 15% of cutaneous melanomas, respectively. Compared to mutant BRAF and NRAS, the role of NF1 loss in melanoma remains understudied. NF1 has a RAS GTPase-activating protein (GAP) function; however, studies also support NF1 RAS-independent tumor-suppressor functions. Recent reports indicate that patients with NF1 mutant melanoma have high response rates to anti-PD-1 immune checkpoint inhibitors (ICIs) for reasons that are not entirely clear. Here, we present data demonstrating that NF1 interacts with PD-L1. Furthermore, NF1 loss in melanoma lines increases PD-L1 cell surface expression through a RAS-GAP-independent mechanism. Co-culture experiments demonstrate that NF1 depletion in melanoma increases resistance to T cell killing, which can be abrogated with anti-PD-1/PD-L1 ICIs. These results support a model whereby NF1 loss leads to immune evasion through the PD-L1/PD-1 axis, providing support for the examination of anti-PD-1 therapies in other NF1 mutant cancers.
Collapse
Affiliation(s)
- Diana Berry
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Dan Moldoveanu
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - Shivshankari Rajkumar
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Mathieu Lajoie
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada
| | - Tiffany Lin
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Daméhan Tchelougou
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Samridhi Sakthivel
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Itai Sharon
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Antoine Bernard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Sandy Pelletier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Yael Ripstein
- Faculty of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Alan Spatz
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada; Lady Davis Institute, McGill University, Montréal, QC H3T 1E1, Canada
| | - Wilson H Miller
- Lady Davis Institute, McGill University, Montréal, QC H3T 1E1, Canada
| | - Rahima Jamal
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada
| | - Réjean Lapointe
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Montreal Neurological Institute and Hospital, Montréal, QC H3A 2B4, Canada
| | - Anne-Marie Mes-Masson
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Kevin Petrecca
- Montreal Neurological Institute and Hospital, Montréal, QC H3A 2B4, Canada
| | | | | | - Beatrice Wang
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - May Chergui
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | | | - Kevin Watters
- McGill University Health Centre, Montréal, QC H4A 3J1, Canada
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - T Martin Schmeing
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Francis Rodier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Department of Radiology, Radio-Oncology and Nuclear Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Simon Turcotte
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM) and Institut du cancer de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | | | | | - Ian R Watson
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada; McGill University Health Centre, Montréal, QC H4A 3J1, Canada.
| |
Collapse
|
119
|
Withers HG, Matsuzaki J, Long M, Rosario SR, Chodon T, Tsuji T, Koya R, Yan L, Wang J, Keler T, Lele SB, Zsiros E, Lugade A, Hutson A, Blank S, Bhardwaj N, Shrikant P, Liu S, Odunsi K. mTOR inhibition modulates vaccine-induced immune responses to generate memory T cells in patients with solid tumors. J Immunother Cancer 2025; 13:e010408. [PMID: 40132910 PMCID: PMC11956311 DOI: 10.1136/jitc-2024-010408] [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: 08/20/2024] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Perturbation of the mechanistic target of rapamycin (mTOR) pathway can instruct effector versus memory cell fate of tumor antigen-specific T cells in preclinical models. In this study, we sought to understand the impact of rapamycin (sirolimus), an mTOR inhibitor, on reprogramming vaccine-induced T cells to enhance memory responses in patients with solid tumors following completion of their standard therapy. METHODS We conducted three phase I clinical trials employing New York esophageal squamous cell carcinoma-1 (NY-ESO-1) vaccination approaches, with or without schedule-varied rapamycin. T cell phenotypes, functions, and Vβ usage in peripheral blood were analyzed to ask whether rapamycin influenced the generation of vaccine-induced T cells with memory attributes. RESULTS The addition of rapamycin to all vaccination approaches was safe and well tolerated. Immediate (days 1-14 postvaccination) or delayed (days 15-28 postvaccination) administration of rapamycin led to a significant increase in the generation of vaccine-induced NY-ESO-1-specific T cells exhibiting central memory phenotypes (CD45RO+CD45RA- CCR7+). Moreover, delayed administration resulted in a greater than threefold (p=0.025) and eightfold (p=0.005) increase in the frequency of NY-ESO-1-specific CD4+ T and CD8+ T cells respectively at the time of long-term follow-up, compared with its immediate usage. CONCLUSION Our novel finding is that delayed administration of rapamycin to patients during the contraction phase of vaccine-induced antitumor immune responses was particularly effective in increasing the frequency of memory T cells up to 1 year postvaccination in patients with solid tumors. Further studies are warranted to identify the impact of this approach on the durability of clinical remission. TRIAL REGISTRATION NUMBER NCT00803569, NCT01536054, NCT01522820.
Collapse
Affiliation(s)
- Henry G Withers
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Junko Matsuzaki
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois, USA
- UChicago Medicine Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Mark Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Spencer R Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Thinle Chodon
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois, USA
- UChicago Medicine Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Takemasa Tsuji
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois, USA
- UChicago Medicine Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Richard Koya
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois, USA
- UChicago Medicine Comprehensive Cancer Center, Chicago, Illinois, USA
| | - Li Yan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Jianming Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Tibor Keler
- R&D, Celldex Therapeutics, Hampton, New Jersey, USA
| | - Shashikant B Lele
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Emese Zsiros
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Amit Lugade
- Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Alan Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Stephanie Blank
- Department of Gynecologic Oncology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Nina Bhardwaj
- Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, New York, USA
| | - Protul Shrikant
- Department of Immunobiology, The University of Arizona College of Medicine Tucson, Tucson, Arizona, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Kunle Odunsi
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois, USA
- UChicago Medicine Comprehensive Cancer Center, Chicago, Illinois, USA
| |
Collapse
|
120
|
Liu F, Qin Y, Luo W, Ruan X, Lu L, Feng B, Yu J. Construction of a risk model associated with tryptophan metabolism and identification of related molecular subtypes in laryngeal squamous cell carcinoma. Front Genet 2025; 16:1530334. [PMID: 40196225 PMCID: PMC11973366 DOI: 10.3389/fgene.2025.1530334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Catabolic metabolites of tryptophan (Trp) are considered to be important microenvironmental factors by suppressing anti-tumor immune responses in cancers. Nevertheless, the effect of Trp metabolism (Trp metabolism)-related genes Trp metabolism-related genes on laryngeal squamous cell carcinoma (LSCC) progression is not yet clear. So, in this study, the TCGA-LSCC, GSE27020, and 40 TMRGs were extracted via public databases to explore the effects of TMRGs on laryngeal squamous cell carcinoma. Firstly, Weighted Gene Co-expression Network Analysis (WGCNA) was adopted with LSCC samples in TCGA-LSCC to acquire key module, and differentially expressed genes between LSCC and normal samples from TCGA-LSCC were yielded via differential expression analysis. Next, differentially expressed TMRGs (DE-TMRGs) was obtained in key model and DEGs, and prognostic genes were identifde through multiple algorithms. Five prognostic genes, namely SERPINA1, TMC8, RENBP, SDS and FAM107A were finally identified. A risk model was established based on the expressions of prognostic genes and survival information of LSCC samples while that were divided into high and low risk groups. Obviously, the LSCC immune dysfunction and exclusion score of high-risk patients was dramatically higher than that in low-risk patients, indicating that patients in the high-risk subgroup exhibited reduced responsiveness to immunotherapy. Besides, the drug sensitivity analysis showed that the low -risk subgroup was notably sensitive to Salubrinal, Lenalidomide, Metformin, while high -risk subgroup was more responsive to Docetaxel, AUY922, Embelin. Eventually, two clusters of LSCC samples had notable correlations with LSCC prognosis. The above results indicated that the risk model consisted of TMRGs (SERPINA1, TMC8, RENBP, SDS and FAM107A) was constructed in LSCC, contributing to studies related to the prognosis and treatment of LSCC.
Collapse
Affiliation(s)
- Feng Liu
- Department of Head and Neck Surgery, Shanxi Cancer Hospital, Taiyuan, China
| | - Yanchao Qin
- Department of Head and Neck Surgery, Shanxi Cancer Hospital, Taiyuan, China
| | - Wei Luo
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - XianHui Ruan
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Lifang Lu
- Department of Head and Neck Surgery, Shanxi Cancer Hospital, Taiyuan, China
| | - Bowei Feng
- School of Stomatology,Shanxi Medical University, Taiyuan, China
| | - Jianfei Yu
- Department of Head and neck radiotherapy, Shanxi Cancer Hospital, Taiyuan, China
| |
Collapse
|
121
|
Du B, Geng J, Wu B, Wang H, Luo R, Liu H, Zhang R, Shan F, Liu L, Zhang S. Pipelines for lymphocyte homeostasis maintenance during cancer immunotherapy. Front Immunol 2025; 16:1522417. [PMID: 40196122 PMCID: PMC11973195 DOI: 10.3389/fimmu.2025.1522417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
In general, increasing lymphocyte entry into tumor microenvironment (TME) and limiting their efflux will have a positive effect on the efficacy of immunotherapy. Current studies suggest maintenance lymphocyte homeostasis during cancer immunotherapy through the two pipelines tumor-associated high endothelial venules and lymphatic vessels. Tumor-associated high endothelial venules (TA-HEVs) play a key role in cancer immunotherapy through facilitating lymphocyte trafficking to the tumor. While tumor-associated lymphatic vessels, in contrast, may promote the egress of lymphocytes and restrict their function. Therefore, the two traffic control points might be potential to maintain lymphocyte homeostasis in cancer during immunotherapy. Herein, we highlight the unexpected roles of lymphocyte circulation regulated by the two gateways for through reviewing the biological characters and functions of TA-HEVs and tumor-associated lymphatic vessels in the entry, positioning and exit of lymphocyte cells in TME during anti-tumor immunity.
Collapse
Affiliation(s)
- Bensu Du
- China Medical University, Shenyang, China
| | - Jin Geng
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, China
| | - Bin Wu
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Houru Wang
- Northeast Yucai Foreign Language School, Shenyang, China
| | - Ru Luo
- Jinzhou Medical University, Jinzhou, China
| | | | - Rui Zhang
- China Medical University, Shenyang, China
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, China
| | - Lei Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shuling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
122
|
Liu Y, Xie Y, Chen Y, Duan J, Bao C, Wang J, Feng H, Wang M, Ren Y, Li P, Luo Q, Xu J, Jiang M, Men Y, Wu Y, Li J, Wang G, Lu W. A protease-cleavable liposome for co-delivery of anti-PD-L1 and doxorubicin for colon cancer therapy in mice. Nat Commun 2025; 16:2854. [PMID: 40128211 PMCID: PMC11933685 DOI: 10.1038/s41467-025-57965-6] [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: 06/24/2023] [Accepted: 03/07/2025] [Indexed: 03/26/2025] Open
Abstract
Immune checkpoint blockade therapy using programmed cell death 1 (PD1) or programmed death ligand 1 (PD-L1) has made significant progress in the treatment of advanced cancers, with some patients achieving long-term remission without clinical recurrence. However, only a minority of colon cancer patients respond to the therapy. Here, we report a protease-cleavable anti-PD-L1 antibody liposome, eLipo anti-PD-L1, for enhancing colon cancer therapy. In vivo, eLipo anti-PD-L1 is cleaved by legumain at colon cancer site into pegylated anti-PD-L1 and cancer-homing doxorubicin liposome. Functional assessments show cancer-targeting, legumain-responding, tumor-penetrating, and immune-activating effects, as well as efficacy in treating colon cancer-bearing mice in vivo. Further mechanistic analysis implicates genes related to T cell differentiation and T cell receptor signaling as potential molecular mediators. Lastly, human colorectal cancer tissue evaluations verify expressions of PD-L1 and legumain, hinting a potential translatability. Our study thus suggests that eLipo anti-PD-L1 may be a feasible vector for co-delivery of immunochemotherapy for colon cancer.
Collapse
Affiliation(s)
- Yixuan Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Ying Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yuling Chen
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jialun Duan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Chunjie Bao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jinling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Hexuan Feng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Mengjie Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yuxin Ren
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Peishan Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Qian Luo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jiarui Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Min Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yanchen Men
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Yang Wu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Jianwei Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Guiling Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Wanliang Lu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China.
- Beijing Key Laboratory of Molecular Pharmaceutics and Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China.
| |
Collapse
|
123
|
Ren X, Guo A, Geng J, Chen Y, Wang X, Zhou L, Shi L. Pan-cancer analysis of co-inhibitory molecules revealing their potential prognostic and clinical values in immunotherapy. Front Immunol 2025; 16:1544104. [PMID: 40196117 PMCID: PMC11973099 DOI: 10.3389/fimmu.2025.1544104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Background The widespread use of immune checkpoint inhibitors (anti-CTLA4 or PD-1) has opened a new chapter in tumor immunotherapy by providing long-term remission for patients. Unfortunately, however, these agents are not universally available and only a minority of patients respond to them. Therefore, there is an urgent need to develop novel therapeutic strategies targeting other co-inhibitory molecules. However, comprehensive information on the expression and prognostic value of co-inhibitory molecules, including co-inhibitory receptors and their ligands, in different cancers is not yet available. Methods We investigated the expression, correlation, and prognostic value of co-inhibitory molecules in different cancer types based on TCGA, UCSC Xena, TIMER, CellMiner datasets. We also examined the associations between the expression of these molecules and the extent of immune cell infiltration. Besides, we conducted a more in-depth study of VISTA. Result The results of differential expression analysis, correlation analysis, and drug sensitivity analysis suggest that CTLA4, PD-1, TIGIT, LAG3, TIM3, NRP1, VISTA, CD80, CD86, PD-L1, PD-L2, PVR, PVRL2, FGL1, LGALS9, HMGB1, SEMA4A, and VEGFA are associated with tumor prognosis and immune cell infiltration. Therefore, we believe that they are hopefully to serve as prognostic biomarkers for certain cancers. In addition, our analysis indicates that VISTA plays a complex role and its expression is related to TMB, MSI, cancer cell stemness, DNA/RNA methylation, and drug sensitivity. Conclusions These co-inhibitory molecules have the potential to serve as prognostic biomarkers and therapeutic targets for a broad spectrum of cancers, given their strong associations with key clinical metrics. Furthermore, the analysis results indicate that VISTA may represent a promising target for cancer therapy.
Collapse
Affiliation(s)
- Xiaoyu Ren
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Anjie Guo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Jiahui Geng
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Yuling Chen
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Xue Wang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Lian Zhou
- Department of Head&Neck Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Lei Shi
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
124
|
Shen X, Yan W, Zhang E, Zhang Z, Zhang Z, Dong H. Adjuvant PD-1 inhibitors improve recurrence and survival outcomes in high-risk hepatocellular carcinoma patients after curative hepatectomy. Eur J Med Res 2025; 30:196. [PMID: 40119430 PMCID: PMC11929280 DOI: 10.1186/s40001-025-02444-3] [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: 09/18/2024] [Accepted: 03/10/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most prevalent malignancy in China, with liver resection recognized as the primary curative intervention. However, HCC patients face an elevated risk of recurrence, thereby significantly impacting prognosis. PURPOSE This study aimed to assess the impact of adjuvant programmed cell death protein-1 (PD-1) inhibitors on survival outcomes in patients with HCC who are at high risk for postoperative recurrence following curative hepatectomy. MATERIALS AND METHODS Among the 199 study participants, 77 received adjuvant PD-1 inhibitors. Propensity score matching (PSM) was used to balance baseline differences between patients who received adjuvant PD-1 inhibitors and those who did not. Assessment of overall survival (OS) and recurrence-free survival (RFS) was conducted using Kaplan-Meier curves, while Cox regression analysis was employed to identify prognostic factors influencing survival. RESULTS After PSM, the 1-year and 2-year RFS were 87.1% and 74.2% in the PD-1 inhibitors group and 44.6% and 37.8% in non-PD-1 inhibitors group (p < 0.001). The 1-year and 2-year OS were 98.5% and 95.7% in the PD-1 inhibitors group compared with 90.7% and 77.0% in non-PD-1 inhibitors group (p = 0.004). Multivariable analyses demonstrated that the use of adjuvant PD-1 inhibitors was significantly associated with improved RFS and OS. Subgroup analysis indicated that adjuvant PD-1 inhibitors group achieved longer RFS than the non-PD-1 inhibitors group in patients without adjuvant transarterial chemoembolization (TACE). CONCLUSION The administration of adjuvant PD-1 inhibitors may effectively reduce the risk of tumor recurrence and improve survival in HCC patients with high risk of recurrence after curative hepatectomy.
Collapse
Affiliation(s)
- Xuehan Shen
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Yan
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Erlei Zhang
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiwei Zhang
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zunyi Zhang
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Hanhua Dong
- Hubei Province for the Clinical Medicine Research Center of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
125
|
Hu Z, Bao Y, Li X, Li Z, Teng P, Liu G, Wang Z. PD-L1 blockade peptide-functionalized NaGdF 4 nanodots for efficient magnetic resonance imaging-guided immunotherapy for breast cancer. RSC Adv 2025; 15:9027-9033. [PMID: 40134683 PMCID: PMC11934231 DOI: 10.1039/d4ra08800j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/09/2025] [Indexed: 03/27/2025] Open
Abstract
Immune checkpoint blockade (ICB) inhibitors have shown great promise for the treatment of numerous types of cancers, including triple-negative breast cancer (TNBC), by interrupting immunosuppressive checkpoints. Herein, programmed cell death ligand 1 (PD-L1) blockade peptide-functionalized NaGdF4 nanodots (designated as PDL1-NaGdF4 NDs) were prepared for magnetic resonance imaging (MRI)-guided TNBC immunotherapy through covalent conjugation of the PD-L1 blockade peptide (sequence, CALNNCVRARTR) with tryptone-capped NaGdF4 NDs (designated as Try-NaGdF4 NDs). MDA-MB-231 tumor could be easily tracked using in vivo MRI with PDL1-NaGdF4 ND enhancement because the as-prepared PDL1-NaGdF4 NDs have a high longitudinal relaxivity (r 1) value (22.8 mM-1 S-1) and accumulate in the tumor site through binding with programmed cell death ligand-1 (PD-L1)-overexpressed cells. A series of in vitro/in vivo results demonstrated that the PDL1-NaGdF4 NDs could effectively suppress MDA-MB-231 tumor growth in mice (66% volume ratio) by inhibiting migration and proliferation of tumor cells. In addition, the results of pharmacokinetic study showed that the PDL1-NaGdF4 NDs were excreted from the body through the kidneys. These results highlight the potential of PDL1-NaGdF4 NDs as a biocompatible nanomedicine for TNBC diagnosis and immunotherapy.
Collapse
Affiliation(s)
- Zhenzhen Hu
- Department of Radiology, China-Japan Union Hospital of Jilin University Changchun 130033 P. R. China
| | - Yunkai Bao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun 130022 P. R. China
| | - Xiaodong Li
- Department of Radiology, China-Japan Union Hospital of Jilin University Changchun 130033 P. R. China
| | - Zhuheng Li
- Jilin Provincial Institute of Education Changchun 130024 P. R. China
| | - Peihong Teng
- Department of Radiology, China-Japan Union Hospital of Jilin University Changchun 130033 P. R. China
| | - Guifeng Liu
- Department of Radiology, China-Japan Union Hospital of Jilin University Changchun 130033 P. R. China
| | - Zhenxin Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences Changchun 130022 P. R. China
| |
Collapse
|
126
|
Thiruppathi J, Vijayan V, Hwang HS, Bang YJ, Loeurng V, Hong SH, Sundaram A, Park IK, Lee SE, Rhee JH. Thermoresistant flagellin-adjuvanted cancer vaccine combined with photothermal therapy synergizes with anti-PD-1 treatment. J Immunother Cancer 2025; 13:e010272. [PMID: 40118497 PMCID: PMC11931959 DOI: 10.1136/jitc-2024-010272] [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: 08/02/2024] [Accepted: 03/04/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Cancer immunotherapy, leveraging the immune system to target and eradicate cancer cells, has transformed cancer treatment paradigms. Immune checkpoint inhibitors (ICIs) are used in a wide array of cancers, but only a limited fraction of patients are responding. Cancer vaccines could elicit antigen-specific immune responses and establish long-term immune memory, preventing recurrence and metastasis. Despite their promising profiles, ICIs and cancer vaccines by themselves are often insufficient to overcome the immunosuppressive tumor microenvironment (TME) and recurrence/metastasis. Addressing these challenges is crucial for improving cancer immunotherapy outcomes. METHODS The targeted liposomal formulation (TLIF), displaying Cyclic RGD (cRGD) peptide on the surface and encapsulating ICG and thermoresistant flagellin (FlaB) inside, was used for photothermal therapy (PTT), which was designed to induce robust immunogenic cell death (ICD) and release tumor antigens (TAs). We employed a mouse breast cancer model amenable to PTT. Utilizing a bilateral DD-Her2/neu tumor implantation model, we evaluated local and abscopal effects of combinatorial approaches employing PTT, FlaB-adjuvanted peptide vaccine (FlaB-Vax), and anti-PD-1 treatment. FlaB-Vax was designed to trigger tumor-associated antigen (TAA)-specific immune responses, which will trigger specific anti-tumor immunity. TLIF-PTT aimed to reduce tumor burden and induce ICD-mediated TA liberation for epitope spreading. Sustained anti-tumor immune memory was assessed by orthotopic rechallenging cured mice with the DD-Her2/neu tumor cells. RESULTS The combination of TLIF-PTT and FlaB-Vax provided significantly enhanced primary tumor suppression, with strong abscopal effects and long-lasting immune memory. The addition of anti-PD-1 therapy further improved long-term relapse-free survival, highlighting the potential of this combinatorial approach to induce durable antitumor immunity and sustainably prevent cancer recurrence and metastasis. CONCLUSION This study demonstrates that the combination of TLIF-PTT and FlaB-Vax synergistically induced synergistic anti-tumor immune responses, which were efficaciously potentiated by anti-PD-1 treatment for recurrence-free long-term survival.
Collapse
Affiliation(s)
| | - Veena Vijayan
- Chonnam National University, Hwasun, Korea (the Republic of)
| | - Hye Suk Hwang
- Chonnam National University, Hwasun, Korea (the Republic of)
- Department of Biomedical Science,College of Life Science and Industry, Sunchon National University, Sunchon 57922, South Korea
| | - Yong Jun Bang
- Chonnam National University, Hwasun, Korea (the Republic of)
| | - Vandara Loeurng
- Chonnam National University, Hwasun, Korea (the Republic of)
| | - Seol Hee Hong
- Chonnam National University, Gwangju, Korea (the Republic of)
| | | | - In-Kyu Park
- Chonnam National University Medical School, Gwangju, Korea (the Republic of)
| | - Shee Eun Lee
- Chonnam National University, Gwangju, Korea (the Republic of)
| | - Joon Haeng Rhee
- Chonnam National University, Hwasun, Korea (the Republic of)
| |
Collapse
|
127
|
Huo W, Chen M, Chang C, Yu J, Chen D, Wang R. Modulation of the tumor immune microenvironment by Interferon Regulatory Factor 8 enhances immunotherapy in lung adenocarcinoma. Sci Rep 2025; 15:9565. [PMID: 40113982 PMCID: PMC11926069 DOI: 10.1038/s41598-025-94424-0] [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: 11/06/2024] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Interferon regulatory factors (IRFs) are integral in governing the expression of Type I interferon (IFN) genes. However, the precise role of IRFs in lung adenocarcinoma remains elusive. Our objective is to elucidate the prognostic implications of IRFs and their potential influence on the immunotherapeutic response in patients with lung adenocarcinoma (LUAD). The association between IRFs expression and clinical as well as prognostic features was evaluated utilizing the TCGA database. Prognostic determinants for LUAD were pinpointed via univariate and multivariate analyses. Nomogram to evaluate prognosis predicated on IRF expression levels. Gene enrichments were conducted to elucidate the mechanisms of action. The degree of immune infiltration was using bioinformatics methods and was validated through a single-cell dataset. We compiled our unique cohort of LUAD patients who underwent anti-PD-1 therapy for subsequent immunohistochemistry and multicolor immunofluorescence staining to gauge the conclusion above. Our findings revealed that IRF8 serves as an independent risk factor for overall survival (OS) in patients with LUAD. An analysis of patients undergoing immunotherapy revealed a positive association between the expression of IRF8 and the response to the treatment. In our specific cohort treated with anti-PD-1, high IRF8 expression was observed to enhance immunotherapy response and prolong OS by modulating immune cell infiltration. Our retrospective analysis suggests that elevated IRF8 expression correlates with improved prognosis in LUAD, with higher IRF8 expression being predictive of a more robust immunotherapy response. Mechanistically, IRF8 expression is associated with a modulated tumor immune microenvironment and improved immunotherapeutic response.
Collapse
Affiliation(s)
- Wen Huo
- Department of Radiation Oncology, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Minxin Chen
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China
| | - Cheng Chang
- Nuclear Medicine Department, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Jinming Yu
- Department of Radiation Oncology, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China.
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| | - Dawei Chen
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, Shandong, China.
| | - Ruozheng Wang
- Department of Radiation Oncology, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China.
| |
Collapse
|
128
|
Zur RT, Zurinam SD, Radman M, Funaro Balouka E, Borodianskiy-Shteinberg T, Saur D, Cohen CJ. Hexokinase2-engineered T cells display increased anti-tumor function. Front Immunol 2025; 16:1477929. [PMID: 40181966 PMCID: PMC11965122 DOI: 10.3389/fimmu.2025.1477929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/11/2025] [Indexed: 04/05/2025] Open
Abstract
Background T cells face significant metabolic challenges in the tumor microenvironment (TME), where cancer cells monopolize critical nutrients like glucose and amino acids. This metabolic competition supports tumor growth while impairing T-cell anti-tumor responses, partly by reducing glycolytic function. Hexokinase 2 (HK2), a key enzyme in glycolysis, plays a pivotal role in maintaining T-cell functionality. Methods To enhance T-cell function, primary human T cells were genetically engineered to overexpress HK2 alongside a tumor-specific receptor. These engineered T cells were tested in vitro and in vivo to evaluate their metabolic and therapeutic efficacy. Results HK2-engineered T cells exhibited increased glycolytic capacity, leading to enhanced cytokine secretion, activation marker expression, and metabolic activity compared to controls. In vivo studies using a human tumor xenograft model demonstrated the superior therapeutic efficacy of HK2-engineered T cells, including delayed tumor growth and improved survival. Conclusion HK2 overexpression improves T-cell metabolic fitness and functionality in hostile TMEs, offering a promising foundation for the development of next-generation immunotherapies targeting T-cell metabolism.
Collapse
Affiliation(s)
- Raphaëlle Toledano Zur
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Shiran Didi Zurinam
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Maria Radman
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Elia Funaro Balouka
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Tatiana Borodianskiy-Shteinberg
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Dieter Saur
- Division of Translational Cancer Research, German Cancer Research Center and German Cancer Consortium, Heidelberg and Center for Translational Cancer Research (TranslaTUM), Institute of Experimental Cancer Therapy, Klinikum Rechts der Isar, School of Medicine, Technische Universität München, Munich, Germany
| | - Cyrille J. Cohen
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
129
|
Hofer TP, Nieto AE, Käsmann L, Pelikan CJ, Taugner J, Mathur S, Eze C, Belka C, Manapov F, Noessner E. Early recovery of leukocyte subsets is associated with favorable progression-free survival in patients with inoperable stage II/III NSCLC after multimodal treatment: a prospective explorative study. Radiat Oncol 2025; 20:43. [PMID: 40114261 PMCID: PMC11927295 DOI: 10.1186/s13014-025-02620-z] [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: 06/21/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND We explored the dynamic changes of major leukocyte subsets during definitive treatment of patients with inoperable stage II/III NSCLC lung cancer and correlated it to survival to identify subpopulations associated with maximal patient benefit. METHODS We analyzed peripheral blood of 20 patients, either treated with thoracic radiotherapy (RT), concurrent chemo-radiotherapy (cCRT), or cCRT with additional immune-checkpoint inhibition therapy. Peripheral blood of 20 patients was collected at 9 timepoints before, during, and up to 1 year post treatment and analyzed by multi-color flow cytometry. Statistical analysis was conducted for leukocyte subpopulations, IL-6, progression-free survival (PFS) and overall survival (OS). RESULTS Increase of absolute lymphocyte counts (ALC) after the end of RT until 6 months thereafter was a predictor of PFS. Baseline lymphocyte counts showed no significant correlation to PFS or OS. Early recovery of absolute counts (AC) at 3 weeks after RT, total CD3 + T-cells, and CD8 + cytotoxic T-cells distinguished those patients with favorable PFS (≥ 12 months) from all other patients. Discriminant analysis identified B-cells, neutrophil-lymphocyte-ratio (NLR), CD4 + T-helper-cells, and NK-cells as predictors of favorable PFS. High variability in IL-6 plasma concentration of consecutive measurements within 6 months after the end of RT correlated negatively with PFS. CONCLUSION Our results suggest that two parameters commonly assessed in clinical routine can be used to predict patient outcome. These are: early increase in CD8 + T-cell lymphocyte count and variability in IL-6 plasma concentration, that are correlated to patients with favorable, respectively, poor outcome after definitive therapy independent of treatment regimen.
Collapse
Affiliation(s)
- Thomas P Hofer
- Helmholtz Zentrum München, Immunoanalytics - Tissue Control of Immunocytes, Munich, Germany.
| | - Alexander E Nieto
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lukas Käsmann
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Comprehensive Cancer Center, Munich, Germany
| | - Carolyn J Pelikan
- Helmholtz Zentrum München, Immunoanalytics - Tissue Control of Immunocytes, Munich, Germany
| | - Julian Taugner
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Saloni Mathur
- Helmholtz Zentrum München, Immunoanalytics - Tissue Control of Immunocytes, Munich, Germany
| | - Chukwuka Eze
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- Comprehensive Cancer Center, Munich, Germany
| | - Farkhad Manapov
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Comprehensive Cancer Center, Munich, Germany
| | - Elfriede Noessner
- Helmholtz Zentrum München, Immunoanalytics - Tissue Control of Immunocytes, Munich, Germany
| |
Collapse
|
130
|
Liu Y, Liu Y, Niu X, Chen A, Li Y, Yu Y, Mo B, Liu Z, Xu T, Cheng J, Wu Z, Wei W. Massively parallel interrogation of human functional variants modulating cancer immunosurveillance. Signal Transduct Target Ther 2025; 10:88. [PMID: 40102418 PMCID: PMC11920242 DOI: 10.1038/s41392-025-02171-5] [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: 06/21/2024] [Revised: 01/23/2025] [Accepted: 02/12/2025] [Indexed: 03/20/2025] Open
Abstract
Anti-PD-1/PD-L1 immune checkpoint blockade (ICB) therapy has revolutionized clinical cancer treatment, while abnormal PD-L1 or HLA-I expression in patients can significantly impact the therapeutic efficacy. Somatic mutations in cancer cells that modulate these critical regulators are closely associated with tumor progression and ICB response. However, a systematic interpretation of cancer immune-related mutations is still lacking. Here, we harnessed the ABEmax system to establish a large-scale sgRNA library encompassing approximately 820,000 sgRNAs that target all feasible serine/threonine/tyrosine residues across the human genome, which systematically unveiled thousands of novel mutations that decrease or augment PD-L1 or HLA-I expression. Beyond residues associated with phosphorylation events, our screens also identified functional mutations that affect mRNA or protein stability, DNA binding capacity, protein-protein interactions, and enzymatic catalytic activity, leading to either gene inactivation or activation. Notably, we uncovered certain mutations that concurrently modulate PD-L1 and HLA-I expression, represented by the clinically relevant mutation SETD2_Y1666. We demonstrated that this mutation induces consistent phenotypic effects across multiple cancer cell lines and enhances the efficacy of immunotherapy in different tumor models. Our findings provide an unprecedented resource of functional residues that regulate cancer immunosurveillance, offering valuable guidance for clinical diagnosis, ICB therapy, and the development of innovative drugs for cancer treatment.
Collapse
Affiliation(s)
- Ying Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Yongshuo Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
- Department of Clinical Laboratory, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xuran Niu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Ang Chen
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yizhou Li
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Ying Yu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Binrui Mo
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Zhiheng Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Tao Xu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Jie Cheng
- Department of pathology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zeguang Wu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China
| | - Wensheng Wei
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Gene Function and Modulation Research, School of Life Sciences, Peking University, Beijing, China.
- Changping Laboratory, Beijing, China.
| |
Collapse
|
131
|
Wen N, Lu Y, Zhuo Y, Fu B, Wang H, He Y, Wu H, Wang Z, Tan W, Qiu L. Enhancing T-Cell Infiltration and Immunity in Solid Tumors via DNA Nanolinker-Mediated Monocyte Hitchhiking. J Am Chem Soc 2025; 147:9800-9809. [PMID: 40042588 DOI: 10.1021/jacs.4c18455] [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: 03/20/2025]
Abstract
Cytotoxic CD8+ T cells are one of the most powerful effectors in the antitumor immune response. However, their insufficient tumor infiltration severely limits the clinical success of immunotherapy in solid tumors. In this work, by using amphiphilic aptamer-incorporated DNA tetrahedra (aptTDN) as the intercellular nanolinker, we developed a monocyte-hitchhiked T-cell delivery strategy to actively promote the intratumoral infiltration of effector CD8+ T cells. Our results demonstrated that membrane-anchoring of aptTDN enabled the specific and stable ligation of T cells with Ly6c+ monocytes, without compromising the migratory behavior of monocytes and the antitumor activity of T cells. By leveraging the intrinsic tumor-homing capability of monocytes, the ligated T cells efficiently accumulated within tumor-associated vasculature and then deeply infiltrated the tumor bed. Additionally, the enhanced intratumoral presence of adoptively transferred effector CD8+ T cells facilitated the establishment of an immunosupportive microenvironment, that further recruited endogenous T cells and ultimately bolstered antitumor immunity. Moreover, our monocyte-hitchhiked T-cell tumor infiltration system could significantly improve the efficacy of immune checkpoint blockade therapy. Collectively, by utilizing chemically synthetic nanolinkers to modulate cellular interactions and develop a delivery system of therapeutic cells, our work presents a new paradigm for the advancement of immunotherapy against solid tumors.
Collapse
Affiliation(s)
- Nachuan Wen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yao Lu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yuting Zhuo
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Bo Fu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Haiyuan Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Yao He
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Hui Wu
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Zhimin Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Liping Qiu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan 410082, China
- The Key Laboratory of Zhejiang Province for Aptamers and Theranostics, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
132
|
Zhang B, Li M, Ji J, Si X, Yin X, Ji G, Ren L, Yao H. A syringeable immunotherapeutic hydrogel enhances T cell immunity via in-situ activation of STING pathway for advanced breast cancer postoperative therapy. Front Immunol 2025; 16:1523436. [PMID: 40176815 PMCID: PMC11961417 DOI: 10.3389/fimmu.2025.1523436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/26/2025] [Indexed: 04/04/2025] Open
Abstract
Complete surgical resection of advanced breast cancer is highly challenging and often leaves behind microscopic tumor foci, leading to inevitable relapse. Postoperative formation of the immunosuppressive tumor microenvironment (TME) reduces the efficacy of immunotherapies against residual tumors. Although cytotoxic chemotherapeutics exert the capacity to intensify cancer immunotherapy via immunogenic cell death (ICD) effects, systemically administered chemo agents often cannot access residual tumor sites, and fail to elicit antitumor immune responses. Herein, we present a novel syringeable immunotherapeutic hydrogel (SiGel@SN38/aOX40) loaded with the DNA-targeting chemotherapeutic 7-ethyl-10-hydroxycamptothecin (SN38) and the anti-OX40 agonist antibody (aOX40). The sustained in-site release of SN38 and aOX40 activate the stimulator of interferon genes (STING) pathway, intensify type I interferons expression, synergistically facilitate dendritic cell (DC) activation, and initiate persistent T cell mediated immune responses within the surgical resection bed that eliminate residual tumors with no tumor recurrence in 120 days. Collectively, our designed SiGel@SN38/aOX40 induces robust and long-lasting tumoricidal immunity following breast cancer resection and exhibit immense potential for clinical translation.
Collapse
Affiliation(s)
- Baozhen Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Min Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Jiahua Ji
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Xiaojiao Yin
- Department of Gynecologic Oncology, Gynecology and Obstetrics Center, the First Hospital of Jilin University, Changchun, China
| | - Guofeng Ji
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Haochen Yao
- Hepatobiliary and Pancreatic Surgery Department, General Surgery Center, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
133
|
Liu YT, Liu HM, Ren JG, Zhang W, Wang XX, Yu ZL, Fu QY, Xiong XP, Jia J, Liu B, Chen G. Immune-featured stromal niches associate with response to neoadjuvant immunotherapy in oral squamous cell carcinoma. Cell Rep Med 2025; 6:102024. [PMID: 40107247 PMCID: PMC11970382 DOI: 10.1016/j.xcrm.2025.102024] [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: 05/05/2024] [Revised: 11/19/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Tumor stromal cells (TSCs) play a crucial yet underexplored role in the tumor microenvironment (TME). This study uses single-cell sequencing and spatial transcriptomics on paired tumor specimens from 22 patients with oral squamous cell carcinoma (OSCC) enrolled in a randomized two-arm phase 2 trial, receiving neoadjuvant anti-PD-1 mono-immunotherapy or anti-PD-1 plus docetaxel-cisplatin-5-fluorouracil (TPF) immunochemotherapy. Single-cell analysis reveals increased TSCs within the TME of responders in immunochemotherapy. Notably, significant post-treatment upregulation of SELP+ high endothelial venules (HEVs) and APOD+ myofibroblastic cancer-associated fibroblasts (myCAFs), alongside a decline in STMN1+ capillary endothelial cells (cECs), is specific to the immunochemotherapy cohort. In contrast, MYF5+ muscle satellite cells (MSCs) are upregulated in non-responders to mono-immunotherapy. SELP+ HEVs and APOD+ myCAFs foster favorable immunomodulatory stromal niches for improved outcomes, while STMN1+ cECs and MYF5+ MSCs form immunosuppressive niches in tumor invasion regions, highlighting therapeutic targets. The trial was registered at ClinicalTrials.gov, and the registration number is NCT04649476.
Collapse
Affiliation(s)
- 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
| | - 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
| | - Jian-Gang Ren
- 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; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Zhang
- 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; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xin-Xin Wang
- 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
| | - Zi-Li Yu
- 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; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Qiu-Yun Fu
- 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; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xue-Peng Xiong
- 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; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jun Jia
- 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; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing 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; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, 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; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China.
| |
Collapse
|
134
|
Kavran AJ, Bai Y, Rabe B, Kreshock A, Fisher A, Cheng Y, Lewin A, Dai C, Meyer MJ, Mavrakis KJ, Lyubetskaya A, Drokhlyansky E. Spatial genomics reveals cholesterol metabolism as a key factor in colorectal cancer immunotherapy resistance. Front Oncol 2025; 15:1549237. [PMID: 40171265 PMCID: PMC11959564 DOI: 10.3389/fonc.2025.1549237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/24/2025] [Indexed: 04/03/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment landscape across multiple cancer types achieving durable responses for a significant number of patients. Despite their success, many patients still fail to respond to ICIs or develop resistance soon after treatment. We sought to identify early treatment features associated with ICI outcome. We leveraged the MC38 syngeneic tumor model because it has variable response to ICI therapy driven by tumor intrinsic heterogeneity. ICI response was assessed based on the level of immune cell infiltration into the tumor - a well-established clinical hallmark of ICI response. We generated a spatial atlas of 48,636 transcriptome-wide spots across 16 tumors using spatial transcriptomics; given the tumors were difficult to profile, we developed an enhanced transcriptome capture protocol yielding high quality spatial data. In total, we identified 8 tumor cell subsets (e.g., proliferative, inflamed, and vascularized) and 4 stroma subsets (e.g., immune and fibroblast). Each tumor had orthogonal histology and bulk-RNA sequencing data, which served to validate and benchmark observations from the spatial data. Our spatial atlas revealed that increased tumor cell cholesterol regulation, synthesis, and transport were associated with a lack of ICI response. Conversely, inflammation and T cell infiltration were associated with response. We further leveraged spatially aware gene expression analysis, to demonstrate that high cholesterol synthesis by tumor cells was associated with cytotoxic CD8 T cell exclusion. Finally, we demonstrate that bulk RNA-sequencing was able to detect immune correlates of response but lacked the sensitivity to detect cholesterol synthesis as a feature of resistance.
Collapse
Affiliation(s)
- Andrew J. Kavran
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Yulong Bai
- Informatics and Predictive Sciences, Bristol Myers Squibb, Cambridge, MA, United States
| | - Brian Rabe
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Anna Kreshock
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Andrew Fisher
- Informatics and Predictive Sciences, Bristol Myers Squibb, Cambridge, MA, United States
| | - Yelena Cheng
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Anne Lewin
- Translational Medicine, Bristol Myers Squibb, Cambridge, MA, United States
| | - Chao Dai
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Matthew J. Meyer
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Konstantinos J. Mavrakis
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Anna Lyubetskaya
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| | - Eugene Drokhlyansky
- Mechanisms of Cancer Resistance Thematic Research Center (TRC), Bristol Myers Squibb, Cambridge, MA, United States
| |
Collapse
|
135
|
Cao LM, Yu YF, Li ZZ, Wang GR, Xiao Y, Luo HY, Liu B, Bu LL. Neoadjuvant Chemoimmunotherapy for Resectable Head and Neck Squamous Cell Carcinoma: Systematic Review and Meta-analysis. Ann Surg Oncol 2025:10.1245/s10434-025-17195-y. [PMID: 40102288 DOI: 10.1245/s10434-025-17195-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Neoadjuvant chemoimmunotherapy provides a good pathological response in patients with resectable head and neck squamous cell carcinoma (HNSCC). Currently, there is no comprehensive systematic review that rigorously evaluates and summarizes the existing studies. In this study, we aimed to synthesize the results on the efficacy of neoadjuvant chemoimmunotherapy in resectable HNSCC to obtain higher-level evidence. METHODS The PubMed, Web of Science, Scopus, and Academic Search Complete (EBSCO) databases, along with ClinicalTrials.gov, Google Scholar, and conference abstracts, were comprehensively searched. The publication dates of the literature were limited to January 2015-July 2024. Meta-analysis was performed using a random-effects model. The percentage of major pathological response (MPR), pathological complete response (pCR), and overall disease-free survival (DFS) were synthesized. The odds ratios of a combined positive score (CPS) ≥ 20 for MPR and the diagnostic performance of using radiological objective response to determine MPR were further explored. RESULTS A total of 13 studies with 458 patients who received neoadjuvant chemoimmunotherapy and 443 patients who underwent curative surgery were included. The pooled MPR, pCR, and overall DFS rates were 61%, 37%, and 91%, respectively. The odds ratios of a CPS ≥ 20 for achieving MPR was 2.09 compared with those with a CPS < 20. The sensitivity of using radiological objective response to determine MPR was 0.91 and the specificity was 0.46, with an area under the curve of 0.76. CONCLUSION Neoadjuvant chemoimmunotherapy showed promising results for resectable HNSCC. A CPS ≥ 20 can be used to screen for treatment-sensitive patients, and radiological examinations can be used to detect pathological response. Definitive conclusions require data from longer follow-up periods and controlled studies.
Collapse
Affiliation(s)
- Lei-Ming Cao
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi-Fu Yu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yao Xiao
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Han-Yue Luo
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing Liu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
136
|
Tsurui T, Hosonuma M, Sasaki A, Maruyama Y, Amari Y, Funayama E, Tajima K, Toyoda H, Isobe J, Yamazaki Y, Baba Y, Shida M, Udaka Y, Mura E, Suzuki R, Iriguchi N, Ishiguro T, Hirasawa Y, Ohkuma R, Shimokawa M, Ariizumi H, Kubota Y, Horiike A, Wada S, Kuramasu A, Tsuji M, Kiuchi Y, Tsunoda T, Yoshimura K. Ki-67 expression in anti-programmed cell death protein-1 antibody-bound CD8 + T cells as a predictor of clinical benefit. Discov Oncol 2025; 16:348. [PMID: 40100476 PMCID: PMC11920542 DOI: 10.1007/s12672-025-02060-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
AIMS Developing predictive biomarkers for immune checkpoint inhibitors (ICIs) is important. Programmed cell death protein-1 (PD-1) receptor occupancy by anti-PD-1 antibodies on circulating T cells varies among patients. However, the association between the exhaustion of these antibody-bound T cells and the clinical efficacy of ICIs remains unknown. Therefore, the present study was aimed at evaluating this association. METHODS This prospective cohort study included patients with advanced non-small cell lung cancer (NSCLC) and esophageal squamous cell carcinoma (ESCC) who received pembrolizumab therapy. Peripheral blood samples were collected during the second cycle of chemotherapy. We analyzed the relationship between exhaustion markers in pembrolizumab-bound (PB) T cells and clinical response. RESULTS A total of 21 patients were analyzed, including 12 patients with NSCLC and 9 patients with ESCC. The expression of Ki-67 in PB-CD8+ TCM and TEM was negatively correlated with both clinical response and overall survival. CONCLUSION The expression of Ki-67 of PB-CD8+ TCM and TEM can serve as a predictive biomarker for the clinical benefit of pembrolizumab therapy. Our study suggests that analyzing antibody-bound T cells could be a novel approach to predict the clinical outcomes of PD-1 blockade therapy.
Collapse
Affiliation(s)
- Toshiaki Tsurui
- Division of Medical Pharmacology, Showa University Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Masahiro Hosonuma
- Division of Medical Pharmacology, Showa University Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Aya Sasaki
- Division of Medical Pharmacology, Showa University Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
- Department of Surgery, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Yuuki Maruyama
- Division of Medical Pharmacology, Showa University Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
- Department of Otorhinolaryngology-Head and Neck Surgery, Showa University School of Medicine, Tokyo, Japan
| | - Yasunobu Amari
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
- Division of Clinical Pharmacology, Showa University Graduate School of Medicine, Tokyo, Japan
- Department of Otolaryngology, Showa University Fujigaoka Hospital, Kanagawa, Japan
| | - Eiji Funayama
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
- Division of Pharmacology, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Kohei Tajima
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Hitoshi Toyoda
- Division of Medical Pharmacology, Showa University Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
- Department of Orthopedic Surgery, Showa University School of Medicine, Tokyo, Japan
| | - Junya Isobe
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
- Department of Hospital Pharmaceutics, Showa University School of Pharmacy, Tokyo, Japan
| | - Yoshitaka Yamazaki
- Pharmacological Research Center, Showa University, Tokyo, Japan
- Division of Toxicology, Showa University Graduate School of Pharmacy, Tokyo, Japan
| | - Yuta Baba
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
- Department of Hematology, Showa University Fujigaoka Hospital, Kanagawa, Japan
| | - Midori Shida
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
| | - Yuko Udaka
- Division of Medical Pharmacology, Showa University Graduate School of Medicine, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Emiko Mura
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
| | - Risako Suzuki
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
| | - Nana Iriguchi
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
| | - Tomoyuki Ishiguro
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
| | - Yuya Hirasawa
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
| | - Ryotaro Ohkuma
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
| | - Masahiro Shimokawa
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
| | - Hirotsugu Ariizumi
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
| | - Yutaro Kubota
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Horiike
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
| | - Satoshi Wada
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Atsuo Kuramasu
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan
| | - Mayumi Tsuji
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Yuji Kiuchi
- Division of Medical Pharmacology, Showa University Graduate School of Medicine, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Takuya Tsunoda
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan
| | - Kiyoshi Yoshimura
- Department of Clinical Immuno-Oncology, Clinical Research Institute for Clinical Pharmacology and Therapeutics, Showa University, 6-11-11 Kitakarasuyama, Setagaya-Ku, Tokyo, 157-8577, Japan.
- Department of Medical Oncology, Showa University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
137
|
Zhu Q, Yu C, Chen Y, Luo W, Li M, Zou J, Xiao F, An S, Saiding Q, Tao W, Kong N, Xie T. Dual mRNA nanoparticles strategy for enhanced pancreatic cancer treatment and β-elemene combination therapy. Proc Natl Acad Sci U S A 2025; 122:e2418306122. [PMID: 40067898 PMCID: PMC11929461 DOI: 10.1073/pnas.2418306122] [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: 09/09/2024] [Accepted: 01/27/2025] [Indexed: 03/25/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is notoriously immune-resistant, limiting the clinical efficacy of single-agent immune modulators and thereby necessitating the exploration of multimodal immunotherapy combinations. Traditional approaches combining conventional immune checkpoint inhibitors with neoantigen vaccines have shown some promise in treating PDAC but are often compromised by intratumoral T lymphocyte exhaustion and systemic toxicity. Hence, novel approaches are needed to address these challenges. Herein, we demonstrate that mRNA polymeric nanoparticles encoding anti-PD-1 antibodies in situ at the tumor site enhance the therapeutic efficacy of neoantigen-based mRNA vaccine for PDAC. This mRNA-based, in situ anti-PD-1 antibody production strategy also protects tumor-infiltrating T cells from PD-1 inhibition, potentially reducing the toxicities induced by systemic checkpoint inhibition. Our study may provide an innovative dual mRNA nanoparticle strategy for effective tumor neoantigen immunotherapy, as well as an mRNA cancer combination therapy strategy with other clinically approved drugs (e.g., β-elemene).
Collapse
Affiliation(s)
- Qianru Zhu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| | - Chuao Yu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| | - Yiquan Chen
- Liangzhu Laboratory, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Eye Center of The Second Affliated Hospital, Zhejiang University, Hangzhou, Zhejiang311121, China
| | - Wei Luo
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| | - Meng Li
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| | - Jianhua Zou
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| | - Fan Xiao
- Liangzhu Laboratory, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Eye Center of The Second Affliated Hospital, Zhejiang University, Hangzhou, Zhejiang311121, China
- Center for Nanomedicine and Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Soohwan An
- Center for Nanomedicine and Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Eye Center of The Second Affliated Hospital, Zhejiang University, Hangzhou, Zhejiang311121, China
- Center for Nanomedicine and Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Tian Xie
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| |
Collapse
|
138
|
Ozasa T, Nakajima M, Tsunedomi R, Goto S, Adachi K, Takahashi H, Tamada K, Nagano H. Novel immune drug combination induces tumour microenvironment remodelling and reduces the dosage of anti-PD-1 antibody. Sci Rep 2025; 15:8956. [PMID: 40089538 PMCID: PMC11910518 DOI: 10.1038/s41598-025-87344-6] [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: 08/21/2024] [Accepted: 01/17/2025] [Indexed: 03/17/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) are effective in clinical settings; however, they present immune-related adverse effects and financial burden. Although dose reduction of ICIs may mitigate these limitations, it could compromise therapeutic efficacy. Using two adjuvants (poly(I:C) and LAG-3-Ig) combined with three neoantigen peptides (Comb), we examined whether Comb could enhance the efficacy of reduced dose of αPD-1 monoclonal antibody (RD-αPD-1 mAb), which has limited efficacy. In a murine colorectal cancer model using an MC38 cell line, Comb addition to RD-αPD-1 mAb enhanced treatment efficacy. Analysis of the tumour microenvironment (TME) in mice treated with Comb using flow cytometry and single-cell RNA sequencing revealed decreased macrophages with highly expressing immunosuppressive genes and increased plasmacytoid dendritic cells with highly expressing antigen-presenting genes. A potent infiltration of CD8+ tumour-infiltrating lymphocytes (TILs) with an effector profile was only observed in RD-αPD-1 mAb with Comb. Additionally, single-cell T cell receptor repertoire analysis underscored an oligoclonal expansion of CD8+ TILs following treatment with RD-αPD-1 mAb with Comb. This novel immune drug combination may be a promising strategy for reducing αPD-1 mAb dosage while preserving antitumour efficacy through modulating the TME.
Collapse
Affiliation(s)
- Takahiro Ozasa
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Masao Nakajima
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
- Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan
| | - Shunsuke Goto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keishi Adachi
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hidenori Takahashi
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Koji Tamada
- Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
- Research Institute for Cell Design Medical Science, Yamaguchi University, Yamaguchi, Japan.
| |
Collapse
|
139
|
Li N, Zhang C, Li X, Liu S, Xu Y, Yang X. Targeting B7-H3 in solid tumors: Development and evaluation of novel CAR-T Cell therapy. Immunobiology 2025; 230:152888. [PMID: 40121824 DOI: 10.1016/j.imbio.2025.152888] [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: 12/01/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025]
Abstract
Ovarian and gastric cancers, representative of many solid tumors, remain among the most challenging malignancies to treat due to limited therapeutic options and poor outcomes at advanced stages. Although immunotherapies have revolutionized cancer treatment, their efficacy in solid tumors has been hindered by issues such as antigen heterogeneity and the immunosuppressive tumor microenvironment. This study presents the development and evaluation of third-generation chimeric antigen receptor T (CAR-T) cells targeting B7-H3, an immune checkpoint molecule widely overexpressed in solid tumors. The B7-H3 CAR-T cells exhibited robust and selective cytotoxicity against B7-H3-positive tumor cells, sparing normal tissues. In preclinical animal models, these cells significantly inhibited tumor growth, demonstrating higher targeting specificity and preferential accumulation in tumor sites. These results highlight B7-H3-targeted CAR-T cells as a potential breakthrough in immunotherapy for solid tumors, offering a foundation for future clinical trials to refine their safety and efficacy.
Collapse
Affiliation(s)
- Ning Li
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa 999078, Macao
| | - Chunhua Zhang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa 999078, Macao; The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Xiaoyu Li
- College of Mechanical and Vehicle Engineering, Chongqing University, Chongqing 400030, China
| | - Shufen Liu
- Surgical Intervention Departments, Hengshui People's Hospital, Hebei 053000, China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa 999078, Macao.
| | - Xifei Yang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa 999078, Macao; Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, No. 8, Longyuan Road, Nanshan District, Shenzhen 518055, China..
| |
Collapse
|
140
|
Sawant A, Shi F, Cararo Lopes E, Hu Z, Abdelfattah S, Baul J, Powers JR, Hinrichs CS, Rabinowitz JD, Chan CS, Lattime EC, Ganesan S, White EP. Immune Checkpoint Blockade Delays Cancer Development and Extends Survival in DNA Polymerase Mutator Syndromes. Cancer Res 2025; 85:1130-1144. [PMID: 39786467 PMCID: PMC11907192 DOI: 10.1158/0008-5472.can-24-2589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/01/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Mutations in the exonuclease domains of the replicative nuclear DNA polymerases POLD1 and POLE are associated with increased cancer incidence, elevated tumor mutation burden (TMB), and enhanced response to immune checkpoint blockade (ICB). Although ICB is approved for treatment of several cancers, not all tumors with elevated TMB respond, highlighting the need for a better understanding of how TMB affects tumor biology and subsequently immunotherapy response. To address this, we generated mice with germline and conditional mutations in the exonuclease domains of Pold1 and Pole. Engineered mice with Pold1 and Pole mutator alleles presented with spontaneous cancers, primarily lymphomas, lung cancer, and intestinal tumors, whereas Pold1 mutant mice also developed tail skin carcinomas. These cancers had highly variable tissue type-dependent increased TMB with mutational signatures associated with POLD1 and POLE mutations found in human cancers. The Pold1 mutant tail tumors displayed increased TMB; however, only a subset of established tumors responded to ICB. Similarly, introducing the mutator alleles into mice with lung cancer driven by mutant Kras and Trp53 deletion did not improve survival, whereas passaging these tumor cells in vitro without immune editing and subsequently implanting them into immunocompetent mice caused tumor rejection in vivo. These results demonstrated the efficiency by which cells with antigenic mutations are eliminated in vivo. Finally, ICB treatment of mutator mice earlier, before observable tumors had developed delayed cancer onset, improved survival and selected for tumors without aneuploidy, suggesting the potential of ICB in high-risk individuals for cancer prevention. Significance: Treating high-mutation burden mice with immunotherapy prior to cancer onset significantly improves survival, raising the possibility of utilizing immune checkpoint blockade for cancer prevention, especially in individuals with increased risk.
Collapse
Affiliation(s)
- Akshada Sawant
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey
| | - Fuqian Shi
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
| | | | - Zhixian Hu
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey
| | - Somer Abdelfattah
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
| | - Jennele Baul
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
| | - Jesse R. Powers
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey
| | | | - Joshua D. Rabinowitz
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey
| | - Chang S. Chan
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Edmund C. Lattime
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Shridar Ganesan
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Eileen P. White
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey
- Department of Molecular Biology and Biochemistry, Piscataway, New Jersey
| |
Collapse
|
141
|
Ye H, Liao W, Pan J, Shi Y, Wang Q. Immune checkpoint blockade for cancer therapy: current progress and perspectives. J Zhejiang Univ Sci B 2025; 26:203-226. [PMID: 40082201 PMCID: PMC11906392 DOI: 10.1631/jzus.b2300492] [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: 07/11/2023] [Accepted: 12/05/2023] [Indexed: 03/16/2025]
Abstract
Dysfunction of anti-tumor immune responses is crucial for cancer progression. Immune checkpoint blockade (ICB), which can potentiate T cell responses, is an effective strategy for the normalization of host anti-tumor immunity. In recent years, immune checkpoints, expressed on both tumor cells and immune cells, have been identified; some of them have exhibited potential druggability and have been approved by the US Food and Drug Administration (FDA) for clinical treatment. However, limited responses and immune-related adverse events (irAEs) cannot be ignored. This review outlines the development and applications of ICBs, potential strategies for overcoming resistance, and future directions for ICB-based cancer immunotherapy.
Collapse
Affiliation(s)
- Hongying Ye
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China
| | - Weijie Liao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China
| | - Jiongli Pan
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China
| | - Yin Shi
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China.
| |
Collapse
|
142
|
Takahashi H, Perez-Villarroel P, Falahat R, Mulé JJ. Targeting MARCO in combination with anti-CTLA-4 leads to enhanced melanoma regression and immune cell infiltration via macrophage reprogramming. J Immunother Cancer 2025; 13:e011030. [PMID: 40081947 PMCID: PMC11907082 DOI: 10.1136/jitc-2024-011030] [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: 11/10/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Strategies to improve the therapeutic efficacy of cancer immunotherapy with immune checkpoint inhibitors include targeting additional immunosuppressive compartments in the tumor microenvironment (TME). Inhibitory macrophages (Mφ) can be one of the most abundant immune cells in the TME associated with poor prognosis. However, to date, selective Mφ depletion strategies as a cancer immunotherapy have not been successful in clinical trials. Macrophage Receptor with Collagenous Structure (MARCO) is one of a family of class-A scavenger receptors expressed by Mφ in the TME and is one of the most upregulated transcripts in dendritic cells (DC) following their ex vivo uptake of dead tumor cells. The clinical significance of MARCO expression in the TME is not fully understood. METHODS The therapeutic potential of targeting MARCO by an anti-murine MARCO (ED31, clone ED31) monoclonal antibody, which inhibits ligand-binding to MARCO, was explored in combination with anti-cytotoxic T-lymphocyte associated protein 4 (anti-CTLA-4) or anti-programmed cell death protein-1 (anti-PD-1) in C57BL/6J mice bearing B16F10 or Pan02 tumors. The mechanism by which ED31 impacts the TME was investigated by flow cytometry in the different treatment arms. The contribution of Mφ was assessed by both in vivo depletion and in vitro functional assays. Chemokine production was measured by a bead-based multiplex assay. RESULTS ED31 enhanced antitumor efficacy of anti-CTLA-4, but not of anti-PD-1. Analysis of the TME revealed that adding ED31 to anti-CTLA-4 substantially increased immune cell infiltration, including mature conventional DC recruitment, that was due to a switch to M1-pattern chemokines by Mφ. Mφ depletion completely abrogated both the increase in immune cell infiltration and chemokine production, and abolished the antitumor efficacy of the combination therapy. CONCLUSIONS Targeting MARCO as an additional checkpoint in the TME can offer a strategy to improve the antitumor efficacy of anti-CTLA-4 through a mechanism involving Mφ reprogramming rather than their depletion.
Collapse
Affiliation(s)
| | | | - Rana Falahat
- Immunology, Moffitt Cancer Center, Tampa, Florida, USA
| | - James J Mulé
- Immunology, Moffitt Cancer Center, Tampa, Florida, USA
- Cutaneous Oncology Program, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
143
|
Qi Z, Cheng Y, Wang K, Cai S, Ni X, Wang T, Zhang K, Jiang S, Xiao Y, Zhang X. Discovery, Synthesis, and Activity Evaluation of Novel Small-Molecule Inhibitors Targeting VISTA for Cancer Immunotherapy. J Med Chem 2025; 68:5222-5237. [PMID: 40014385 DOI: 10.1021/acs.jmedchem.4c02031] [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: 03/01/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have been potent therapeutic options for the treatment of multiple types of cancer. However, not all patients experience benefits from ICIs, and discovering inhibitors targeting novel immune checkpoints is necessary. V-domain Ig suppressor of T-cell activation (VISTA) is a novel immune checkpoint. Blockade of the VISTA pathway enhances antitumor immunity in multiple tumor types. Herein, a series of VISTA inhibitors based on the benzimidazole scaffold were discovered. B3 showed the strongest binding affinity to the VISTA protein with a KD value of 0.452 ± 0.12 μM. In vitro, B3 could effectively activate VISTA-mediated immunosuppression and induce effective VISTA degradation in HepG2 cells. In vivo, B3 improved pharmacokinetics compared to the lead compound 4. Moreover, compound B3 significantly inhibited tumor growth in a CT26 colon cancer model. These results suggest that compound B3 is a promising VISTA small molecule inhibitor and degrader worthy of further development as an antitumor agent.
Collapse
Affiliation(s)
- Zhihao Qi
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yao Cheng
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kaizhen Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shi Cai
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiang Ni
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kuojun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Sheng Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yibei Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyu Zhang
- Department of Biomedical Engineering and Diagnostic Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
144
|
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] [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
|
145
|
Matsumoto K, Matsumoto Y, Wada J. PARylation-mediated post-transcriptional modifications in cancer immunity and immunotherapy. Front Immunol 2025; 16:1537615. [PMID: 40134437 PMCID: PMC11933034 DOI: 10.3389/fimmu.2025.1537615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Poly-ADP-ribosylation (PARylation) is a post-translational modification in which ADP-ribose is added to substrate proteins. PARylation is mediated by a superfamily of ADP-ribosyl transferases known as PARPs and influences a wide range of cellular functions, including genome integrity maintenance, and the regulation of proliferation and differentiation. We and others have recently reported that PARylation of SH3 domain-binding protein 2 (3BP2) plays a role in bone metabolism, immune system regulation, and cytokine production. Additionally, PARylation has recently gained attention as a target for cancer treatment. In this review, we provide an overview of PARylation, its involvement in several signaling pathways related to cancer immunity, and the potential of combination therapies with PARP inhibitors and immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Yoshinori Matsumoto
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Faculty of
Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | |
Collapse
|
146
|
Fobian SF, Amin M, Sacchetti A, Oei AL, Ten Hagen TLM. Investigating the delivery of PD-L1-targeted immunoliposomes in a dynamic cervical cancer-on-a-chip model. J Control Release 2025; 379:236-250. [PMID: 39798703 DOI: 10.1016/j.jconrel.2025.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/14/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
The recent approval of pembrolizumab in recurrent or metastatic cervical cancer warrants further investigations into the usefulness of immunotherapies for more durable and less radical interventions. In this study, the targeting potential of anti-PD-L1-functionalized immunoliposomes was tested in a 3D in vitro cervical cancer-on-a-chip model. Immunolipsomes were synthesized and decorated externally with monovalent anti-PD-L1 Fab' fragments of commercially available atezolizumab. Cervical cancer cell lines with varying levels of PD-L1 expression were cultured as spheroids embedded in a collagen I matrix, and treated under flow of culture media. Flow cytometry and live-cell confocal imaging were used to measure the interactions and uptake of untargeted liposomes and immunoliposomes in this panel of cell lines. The immunoliposomes retained specific functionality regardless of protein corona formation in high serum environments. As such, spheroids expressing high levels of PD-L1 preferentially internalized immunoliposomes in a 3D environment with extracellular matrix present, while low PD-L1-expressing cell lines showed no preference for either formulation. Importantly, treatments performed in monolayer cultures (on plastic) showed no differences between immuno- and untargeted liposome uptake, including the way in which the endocytosed liposomes are trafficked subcellularly. This study demonstrates the importance of both active and passive accumulation strategies to achieve nanoparticle targeting. Immunoliposomes remain a promising platform for the development of targeted nanotherapies against cervical cancers. However, initial functional tests did not translate directly to biological performance and this should be kept in mind for future formulations. Furthermore, the in vitro model developed appeared useful for visualizing liposome uptake in a 3D, live tissue environment and represents a cost-effective and reproducible model for future studies.
Collapse
Affiliation(s)
- Seth-Frerich Fobian
- Precision Medicine in Oncology (PrMiO), and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands; Department of Radiation Oncology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Cancer Center Amsterdam (CCA), Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Mohamadreza Amin
- Precision Medicine in Oncology (PrMiO), and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands
| | - Andrea Sacchetti
- Department of Pathology, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Arlene L Oei
- Department of Radiation Oncology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), Cancer Center Amsterdam (CCA), Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Timo L M Ten Hagen
- Precision Medicine in Oncology (PrMiO), and Nanomedicine Innovation Center Erasmus (NICE), Department of Pathology, Erasmus MC Cancer Institute, Erasmus MC, Dr. Molewaterplein 40, 3015 GD Rotterdam, the Netherlands.
| |
Collapse
|
147
|
Zhang W, Liu S, Hou Y, Xu S, An J, Lee K, Miao Q, Wang N, Wang Y, Ma M. Functional nanoplatform for modulating cellular forces to enhance antitumor immunity via mechanotransduction. J Control Release 2025; 379:850-865. [PMID: 39863022 DOI: 10.1016/j.jconrel.2025.01.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/18/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Immune cells are sensitive to the perception of mechanical stimuli in the tumor microenvironment. Changes in biophysical cues within tumor tissue can alter the force-sensing mechanisms experienced by cells. Mechanical stimuli within the extracellular matrix are transformed into biochemical signals through mechanotransduction. Delving into how these minute biophysical cues affect the activation of immune cells, metabolic reprogramming, and subsequent effector functions could offer perspectives on therapeutic interventions for immune-related disorders. Our study used a ternary phycocyanin-podophyllotoxin-IDO1 self-assembled nanoplatform to investigate molecule-scale regulation of mechanical cues in the tumor microenvironment on immune cell functions to modulate immune responses. After treatment, a caspase cascade was mediated by remodeling mechanical cues, including cytoskeleton-related assembly, force channel activation, and metabolic reprogramming, all of which contributed to enhancing anti-tumor immunity via mechanotransduction. The results will be helpful for understanding the interaction between cell force remodeling and antitumor immunity via mechanotransduction.
Collapse
Affiliation(s)
- Wanheng Zhang
- Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China; Department of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shuqin Liu
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yan Hou
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Shihui Xu
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Jiayan An
- School of Life Science, University of Liverpool, Liverpool L69 3BX, UK
| | - Kyubae Lee
- Department of Biomedical Materials, Konyang University, Daejeon 35365, Republic of Korea
| | - Qi Miao
- Department of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Nana Wang
- Department of Pediatrics, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, China.
| | - Yongtao Wang
- School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Mengze Ma
- Medical 3D Printing Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China.
| |
Collapse
|
148
|
Ludwig RJ, Anson M, Zirpel H, Thaci D, Olbrich H, Bieber K, Kridin K, Dempfle A, Curman P, Zhao SS, Alam U. A comprehensive review of methodologies and application to use the real-world data and analytics platform TriNetX. Front Pharmacol 2025; 16:1516126. [PMID: 40129946 PMCID: PMC11931024 DOI: 10.3389/fphar.2025.1516126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/13/2025] [Indexed: 03/26/2025] Open
Abstract
Randomized controlled trials (RCTs) are the gold standard for evaluating the efficacy and safety of both pharmacological and non-pharmacological interventions. However, while they are designed to control confounders and ensure internal validity, their usually stringent inclusion and exclusion criteria often limit the generalizability of findings to broader patient populations. Moreover, RCTs are resource-intensive, frequently underpowered to detect rare adverse events, and sometimes narrowly focused due to their highly controlled environments. In contrast, real-world data (RWD), typically derived from electronic health records (EHRs) and claims databases, offers a valuable counterpart for answering research questions that may be impractical to address through RCTs. Recognizing this, the US Food and Drug Administration (FDA) has increasingly relied on real-world evidence (RWE) from RWD to support regulatory decisions and post-market surveillance. Platforms like TriNetX, that leverage large-scale RWD, facilitate collaborations between academia, industry, and healthcare organizations, and constitute an in-depth tool for retrieval and analysis of RWD. TriNetX's federated network architecture allows real-time, privacy-compliant data access, significantly enhancing the ability to conduct retrospective studies and refine clinical trial designs. With access to currently over 150 million EHRs, TriNetX has proven particularly effective in filling gaps left by RCTs, especially in the context of rare diseases, rare endpoints, and diverse patient populations. As the role of RWD in healthcare continues to expand, TriNetX stands out as a critical tool that complements traditional clinical trials, bridging the gap between controlled research environments and real-world practice. This review provides a comprehensive analysis of the methodologies and applications of the TriNetX platform, highlighting its potential contribution to advance patient care and outcomes.
Collapse
Affiliation(s)
- Ralf J. Ludwig
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Department of Dermatology, University Hospital Schleswig-Holstein Lübeck, Lübeck, Germany
- Institute and Comprehensive Centre for Inflammation Medicine, University-Hospital Schleswig-Holstein, Lübeck, Germany
| | - Matthew Anson
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Medicine, Aintree University Hospital, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
| | - Henner Zirpel
- Institute and Comprehensive Centre for Inflammation Medicine, University-Hospital Schleswig-Holstein, Lübeck, Germany
| | - Diamant Thaci
- Institute and Comprehensive Centre for Inflammation Medicine, University-Hospital Schleswig-Holstein, Lübeck, Germany
| | - Henning Olbrich
- Department of Dermatology, University Hospital Schleswig-Holstein Lübeck, Lübeck, Germany
| | - Katja Bieber
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
| | - Khalaf Kridin
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
- Unit of Dermatology and Skin Research Laboratory, Galilee Medical Center, Nahariya, Israel
| | - Astrid Dempfle
- Institute of Medical Informatics and Statistics, Kiel University, Kiel, Germany
| | - Philip Curman
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Sizheng S. Zhao
- Centre for Musculoskeletal Research at University of Manchester, Manchester, United Kingdom
| | - Uazman Alam
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Medicine, Aintree University Hospital, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
- Centre for Biomechanics and Rehabilitation Technologies, Staffordshire University, Stoke-onTrent, United Kingdom
| |
Collapse
|
149
|
Zhang Y, Chen H, Mo H, Zhao N, Sun X, Liu B, Gao R, Xu B, Zhang Z, Liu Z, Ma F. Distinct cellular mechanisms underlie chemotherapies and PD-L1 blockade combinations in triple-negative breast cancer. Cancer Cell 2025; 43:446-463.e7. [PMID: 39919737 DOI: 10.1016/j.ccell.2025.01.007] [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: 06/01/2024] [Revised: 11/05/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025]
Abstract
Combining immune checkpoint blockade (ICB) with chemotherapy shows promise for treating triple-negative breast cancer (TNBC), though the mechanisms remain incompletely understood. Here, we integrate published and new single-cell RNA sequencing (scRNA-seq) data to investigate the tumor immune microenvironment (TIME) in TNBC patients treated with paclitaxel (PTX), nab-paclitaxel (Nab-PTX), and their combinations with the anti-PD-L1 antibody atezolizumab (ATZ). Compared to ATZ plus PTX, ATZ plus Nab-PTX rewires TCF7+ stem-like effector memory CD8+ T cells (Tsem) and CD4+ T follicular helper (Tfh) cells. Nab-paclitaxel, unlike PTX, also reshapes the myeloid compartment, expanding mast cells and pro-inflammatory macrophages. Our analyses in human TNBC and murine models underscore the crucial role of mast cells in orchestrating anti-tumor immune responses, likely by promoting the recruitment and activation of T and B cells. In vivo experiments demonstrate that activating mast cells alongside PD-L1 blockade attenuates TNBC progression, suggesting mast cells as a promising adjunct for enhancing ICB therapy efficacy.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China.
| | - Hongyan Chen
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hongnan Mo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ning Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xiaoying Sun
- Department of Medical Oncology, Cancer Hospital of HuanXing, ChaoYang District, Beijing 100005, China
| | - Baolin Liu
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ranran Gao
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China
| | - Binghe Xu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Zemin Zhang
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing 100871, China.
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Fei Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China; Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
150
|
Wei JR, Lu MY, Wei TH, Fleishman JS, Yu H, Chen XL, Kong XT, Sun SL, Li NG, Yang Y, Ni HW. Overcoming cancer therapy resistance: From drug innovation to therapeutics. Drug Resist Updat 2025; 81:101229. [PMID: 40081221 DOI: 10.1016/j.drup.2025.101229] [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: 12/26/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
One of the major limitations of cancer therapy is the emergence of drug resistance. This review amis to provide a focused analysis of the multifactorial mechanisms underlying therapy resistance,with an emphasis on actionable insights for developing novel therapeutic strategies. It concisely outlines key factors contributing to therapy resistance, including drug delivery barriers, cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT), cancer heterogeneity, tumor microenvironment (TME), genetic mutations, and alterlations in gene expression. Additionally, we explore how tumors evade targeted therapies through pathway-specific mechanisms that restore disrupted signaling pathways. The review critically evaluates innovative strategies designed to sensitize resistant tumor cells, such as targeted protein dedgradation, antibody-drug conjugates, structure-based drug design, allosteric drugs, multitarget drugs, nanomedicine and others We also highlight the importance of understanding the pharmacological actions of these agents and their integration into treatment regimens. By synthesizing current knowledge and identifying gaps in our understanding, this review aims to guide future research and improve patient outcomes in cancer therapy.
Collapse
Affiliation(s)
- Jin-Rui Wei
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China; The First Clinical College of Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Meng-Yi Lu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Tian-Hua Wei
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Joshua S Fleishman
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Hui Yu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Xiao-Li Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Xiang-Tu Kong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Shan-Liang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China.
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Ye Yang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Hai-Wen Ni
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|