1
|
Wang M, Zhang Z, Yang Y, Peng X, Yin H. A targeted MAVS fusion protein for controlled innate immune activation and antitumor therapy. Oncoimmunology 2025; 14:2478850. [PMID: 40085508 PMCID: PMC11913393 DOI: 10.1080/2162402x.2025.2478850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/25/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Targeted therapies leveraging the innate immune system are emerging as promising cancer treatments. The mitochondrial antiviral signaling protein (MAVS) plays a crucial role in initiating innate immune responses, but its clinical use is limited by the risk of uncontrolled activation and systemic toxicity. To address this, we developed a novel therapeutic agent, the truncated interferon activation switch (TRIAS), combining MAVS truncates with a tumor antigen-targeting single-chain variable fragment (scFv). This design ensures antigen-dependent, controlled activation. Lentiviral delivery of TRIAS induced significant antitumor responses, including complete tumor regression in some cases. Flow cytometry (FCM) analysis further confirmed that tumor cells were the predominant population expressing the transgene. TRIAS-expressing tumor cells exhibited enhanced antitumor activity, likely due to increased cytokine release and upregulated major histocompatibility complex (MHC) expression, enabling tumor cells to function as antigen-presenting cells. This activated other immune cells, driving adaptive immune responses. Additionally, TRIAS promoted a proinflammatory shift in the tumor microenvironment (TME). In conclusion, TRIAS was validated as an innovative immunotherapeutic agent with MAVS-like immune-activating properties and tightly controlled mechanisms, offering a safer and more effective approach for clinical cancer immunotherapy.
Collapse
Affiliation(s)
- Muhan Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhijie Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - YouYou Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaoyi Peng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Hongping Yin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
2
|
Wang W, Zhai Y, Yang X, Ye L, Lu G, Shi X, Zhai G. Effective design of therapeutic nanovaccines based on tumor neoantigens. J Control Release 2025; 380:17-35. [PMID: 39892648 DOI: 10.1016/j.jconrel.2025.01.078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Neoantigen vaccines are among the most potent immunotherapies for personalized cancer treatment. Therapeutic vaccines containing tumor-specific neoantigens that elicit specific T cell responses offer the potential for long-term clinical benefits to cancer patients. Unlike immune-checkpoint inhibitors (ICIs), which rely on pre-existing specific T cell responses, personalized neoantigen vaccines not only promote existing specific T cell responses but importantly stimulate the generation of neoantigen-specific T cells, leading to the establishment of a persistent specific memory T cell pool. The review discusses the current state of clinical research on neoantigen nanovaccines, focusing on the application of vectors, adjuvants, and combinational strategies to address a range of challenges and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Weilin Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States of America
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
3
|
Yang Y, Yu Q, Zhang H, Liu Y, Wang H, Yang N, Shi Y, Zhang W, Wu Z, Huang S, Xie W, Duan R, Mao Q, Shi X, Gao Z, Wang X, Guo H, Chen L, Han Y, Li X, Chen L, Tang S, Fan Y, Yao W, Tian H, Gao X. Restoring tumor antigenicity activates the "bystander" T cell immune cycle. J Control Release 2025; 380:256-268. [PMID: 39894265 DOI: 10.1016/j.jconrel.2025.01.094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Tumor-specific T cells play a crucial role in tumor immunity. However, these cells are often scarce and functionally exhausted within the tumor microenvironment (TME), leading to the limited efficacy of immunotherapy in many cancer patients. In contrast, increasing evidence suggests that the TME is rich in "bystander" T cells (TBYS), most of which are virus-specific and unrelated to the tumor. These TBYS cells retain functional memory characteristics and the potential to kill tumor cells. To utilize TBYS cells in the TME for tumor elimination, we designed an intracellular delivery system, ASCP, encoding a TBYS epitope to redirect tumor cell antigen specificity toward pre-existing TBYS cells, resulting in effective tumor inhibition in multiple preclinical models. The ASCP-antigen peptide strategy restores the antigenicity of tumor cells and induces epitope spreading of tumor antigens, thereby eliciting more diverse tumor-specific T cell responses. Remarkably, this strategy incorporates MHC-II epitopes containing unnatural amino acids (p-nitrophenylalanine, termed NiraTh), which stimulate CD4+ T cell-mediated immunity and assist CD8+ T cells in clearing tumors. Overall, the ASCP-mediated tumor antigen reprogramming strategy provides important insights for cancer immunotherapy in populations with a history of common viral infections.
Collapse
Affiliation(s)
- Yifan Yang
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Qiumin Yu
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Haoyu Zhang
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Yuchen Liu
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Hexuan Wang
- Shenyang Institutes for Food and Drug Control, China
| | - Ningyi Yang
- School of Pharmacy, Nanjing Medical University, China
| | - Yulian Shi
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Wanli Zhang
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Zijie Wu
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Shitong Huang
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Wenbin Xie
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Ran Duan
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Qiuli Mao
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Xupeiyao Shi
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Zheng Gao
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Xiaoning Wang
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Hanlin Guo
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Lingxiao Chen
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Yi Han
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Ximing Li
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Liyuan Chen
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Siqi Tang
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Ying Fan
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China
| | - Wenbing Yao
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China.
| | - Hong Tian
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China.
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Draggability of Biopharmaceuticals and State, Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, China.
| |
Collapse
|
4
|
Li Y, Sun Q, Yang Z, Luo M, Yang W, Song Z, Liu K, Li H, Gao W, Wu Q, Shen W, Yang Y, Yin D. Transforming tumors into 'high-risk bombs' triggers a neoantigen storm and amplifies immune responses. J Control Release 2025; 380:1080-1094. [PMID: 39971250 DOI: 10.1016/j.jconrel.2025.02.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/14/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
Although various immunotherapies have improved the treatment of several challenging malignancies in clinical applications, current research suggests that neoantigens remain fundamental to the initiation of immunotherapy, implying a dependence on high mutation loads in tumors and stable target antigens. To overcome these limitations, we propose a novel immunotherapy paradigm that interferes with splicing to induce the expression of neoantigens and neoepitopes while simultaneously blocking autophagy to prevent their degradation through endogenous pathways. This approach ensures the stable expression and accumulation of neoantigens and neoepitopes in tumor cells. To fully unleash the potential of neoantigens, we further induce tumors to undergo immunogenic cell death (ICD), triggering a "neoantigen storm" at the tumor site to recruit and activate more dendritic cells (DCs). Through a DC-dependent mechanism, communication between the tumor and the tumor-draining lymph node (TDLN) is enhanced, summoning more neoantigen-specific cytotoxic T lymphocytes to lyse tumor cells and establish immune circulation. In summary, this work presents a novel antigen-based immune sensitization strategy that stabilizes target antigens while exploring the potential of non-targeted antigens. By bypassing the cumbersome neoantigen identification process, this strategy holds promise for rapid clinical application in combination with other immunotherapies.
Collapse
Affiliation(s)
- Yunlong Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Quanwei Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Zexin Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Min Luo
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wenshuo Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Zhengwei Song
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Kang Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Huihui Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wenheng Gao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Qinghua Wu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China; Anhui Provincial Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230021, China.
| | - Ye Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230031, China; Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China.
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China; Anhui Provincial Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230021, China; Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei 230012, China.
| |
Collapse
|
5
|
Leung KK, Schaefer K, Lin Z, Yao Z, Wells JA. Engineered Proteins and Chemical Tools to Probe the Cell Surface Proteome. Chem Rev 2025. [PMID: 40178992 DOI: 10.1021/acs.chemrev.4c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
The cell surface proteome, or surfaceome, is the hub for cells to interact and communicate with the outside world. Many disease-associated changes are hard-wired within the surfaceome, yet approved drugs target less than 50 cell surface proteins. In the past decade, the proteomics community has made significant strides in developing new technologies tailored for studying the surfaceome in all its complexity. In this review, we first dive into the unique characteristics and functions of the surfaceome, emphasizing the necessity for specialized labeling, enrichment, and proteomic approaches. An overview of surfaceomics methods is provided, detailing techniques to measure changes in protein expression and how this leads to novel target discovery. Next, we highlight advances in proximity labeling proteomics (PLP), showcasing how various enzymatic and photoaffinity proximity labeling techniques can map protein-protein interactions and membrane protein complexes on the cell surface. We then review the role of extracellular post-translational modifications, focusing on cell surface glycosylation, proteolytic remodeling, and the secretome. Finally, we discuss methods for identifying tumor-specific peptide MHC complexes and how they have shaped therapeutic development. This emerging field of neo-protein epitopes is constantly evolving, where targets are identified at the proteome level and encompass defined disease-associated PTMs, complexes, and dysregulated cellular and tissue locations. Given the functional importance of the surfaceome for biology and therapy, we view surfaceomics as a critical piece of this quest for neo-epitope target discovery.
Collapse
Affiliation(s)
- Kevin K Leung
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, United States
| | - Kaitlin Schaefer
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, United States
| | - Zhi Lin
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, United States
| | - Zi Yao
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, United States
| | - James A Wells
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California 94158, United States
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
6
|
Ding YP, Liu CC, Yu KD. RNA modifications in the tumor microenvironment: insights into the cancer-immunity cycle and beyond. Exp Hematol Oncol 2025; 14:48. [PMID: 40176140 PMCID: PMC11963313 DOI: 10.1186/s40164-025-00648-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025] Open
Abstract
The chemical modification of biological molecules is a critical regulatory mechanism for controlling molecular functions. Although research has long focused on DNA and proteins, RNA modifications have recently attracted substantial interest with the advancement in detection technologies. In oncology, many studies have identified dysregulated RNA modifications including m6A, m1A, m5C, m7G, pseudouridylation and A to I editing, leading to disrupted downstream pathways. As the concept of the tumor microenvironment has gained prominence, studies have increasingly examined the role of RNA modifications in this context, focusing on interactions among cancer cells, immune cells, stromal cells, and other components. Here we review the RNA modifications in the tumor microenvironment through the perspective of the Cancer-Immunity Cycle. The extracellular RNA modifications including exosomes and influence of microbiome in RNA modifications are potential research questions. Additionally, RNA modifying enzymes including FTO, ALKBH5, METTL3, PUS7 are under investigation as potential biomarkers and targets for combination with immunotherapies. ADCs and mimetics of modified RNA could be potential novel drugs. This review discusses the regulatory roles of RNA modifications within the tumor microenvironment.
Collapse
Affiliation(s)
- You-Peng Ding
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Cui-Cui Liu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Ke-Da Yu
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
7
|
Pan Y, Zeng F, Luan X, He G, Qin S, Lu Q, He B, Han X, Song Y. Polyamine-Depleting Hydrogen-Bond Organic Frameworks Unleash Dendritic Cell and T Cell Vigor for Targeted CRISPR/Cas-Assisted Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411886. [PMID: 39972681 DOI: 10.1002/adma.202411886] [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: 08/12/2024] [Revised: 01/26/2025] [Indexed: 02/21/2025]
Abstract
Polyamines have tantalized cancer researchers as a potential means to rein in the rampant growth of cancer cells. However, clinical trials in recent decades have disappointed in delivering notable progress. Herein, a microfluidic-assisted synthetic hydrogen-bond organic framework (HOF) as a polyamine-depleting nanoplatforms designed to unleash the vigor of both dendritic cells (DCs) and T cells for precision cancer immunotherapy is reported. Upon internalization by tumor cells, the loaded plasma amine oxidase (PAO) in HOF efficiently depletes polyamines, remolding the tumor microenvironment and alleviating T-cell immunosuppression. This process also generates acrolein and H2O2, triggering CRISPR-assisted neoantigen generation. Specifically, Acrolein induces carbonyl stress, increasing mutational burdens. Simultaneously, HOF leverages the energy from the bis[2,4,5-trichloro-6-(pentyloxycarbonyl)phenyl] oxalate (CPPO)-H2O2 reaction for CRET-triggered singlet oxygen production, leading to thioether bond cleavage and release CRISPR-Cas9. Once released, CRISPR-Cas9 knocks out the DNA mismatch repair (MMR)-related MLH1 gene, further elevating mutational burdens and generating neoantigens, ideal targets for DCs. This dual-action strategy not only corrects T-cell immunosuppression but also enhances DC efficacy, presenting a powerful approach for tumor immunotherapy.
Collapse
Affiliation(s)
- Yongchun Pan
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, Nanjing University, Nanjing, 210023, China
| | - Fei Zeng
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, Nanjing University, Nanjing, 210023, China
| | - Xiaowei Luan
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, Nanjing University, Nanjing, 210023, China
| | - Guanzhong He
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, Nanjing University, Nanjing, 210023, China
| | - Shurong Qin
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, Nanjing University, Nanjing, 210023, China
| | - Qianglan Lu
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, Nanjing University, Nanjing, 210023, China
| | - Bangshun He
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xin Han
- School of Medicine & Holistic Integrative Medicine, JiangsuCollaborative Innovation Canter of Chinese Medicinal ResourcesIndustrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yujun Song
- College of Engineering and Applied Sciences, State Key Laboratory of Analytical Chemistry for Life Science, National Laboratory of Solid State Microstructures, Nanjing University, Nanjing, 210023, China
| |
Collapse
|
8
|
Ding Y, Yu Y. Therapeutic potential of flavonoids in gastrointestinal cancer: Focus on signaling pathways and improvement strategies (Review). Mol Med Rep 2025; 31:109. [PMID: 40017144 PMCID: PMC11884236 DOI: 10.3892/mmr.2025.13474] [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: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
Flavonoids are a group of polyphenolic compounds distributed in vegetables, fruits and other plants, which have considerable antioxidant, anti‑tumor and anti‑inflammatory activities. Several types of gastrointestinal (GI) cancer are the most common malignant tumors in the world. A large number of studies have shown that flavonoids have inhibitory effects on cancer, and they are recognized as a class of potential anti‑tumor drugs. Therefore, the present review investigated the molecular mechanisms of flavonoids in the treatment of different types of GI cancer and summarized the drug delivery systems commonly used to improve their bioavailability. First, the classification of flavonoids and the therapeutic effects of various flavonoids on human diseases were briefly introduced. Then, to clarify the mechanism of action of flavonoids on different types of GI cancer in the human body, the metabolic process of flavonoids in the human body and the associated signaling pathways causing five common types of GI cancer were discussed, as well as the corresponding therapeutic targets of flavonoids. Finally, in clinical settings, flavonoids have poor water solubility, low permeability and inferior stability, which lead to low absorption efficiency in vivo. Therefore, the three most widely used drug delivery systems were summarized. Suggestions for improving the bioavailability of flavonoids and the focus of the next stage of research were also put forward.
Collapse
Affiliation(s)
- Ye Ding
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yong Yu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
9
|
Linderman SW, DeRidder L, Sanjurjo L, Foote MB, Alonso MJ, Kirtane AR, Langer R, Traverso G. Enhancing immunotherapy with tumour-responsive nanomaterials. Nat Rev Clin Oncol 2025; 22:262-282. [PMID: 40050505 DOI: 10.1038/s41571-025-01000-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 03/09/2025]
Abstract
The targeted delivery of immunotherapies to tumours using tumour-responsive nanomaterials is a promising area of cancer research with the potential to address the limitations of systemic administration such as on-target off-tumour toxicities and a lack of activity owing to the immunosuppressive tumour microenvironment (TME). Attempts to address these challenges include the design and functionalization of nanomaterials capable of releasing their cargoes in response to specific TME characteristics, thus facilitating the targeted delivery of immune-checkpoint inhibitors, cytokines, mRNAs, vaccines and, potentially, chimaeric antigen receptors as well as of agents that modulate the extracellular matrix and induce immunogenic cell death. In this Review, we describe these various research efforts in the context of the dynamic properties of the TME, such as pH, reductive conditions, reactive oxygen species, hypoxia, specific enzymes, high levels of ATP and locoregional aspects, which can be leveraged to enhance the specificity and efficacy of nanomaterial-based immunotherapies. Highlighting preclinical successes and ongoing clinical trials, we evaluate the current landscape and potential of these innovative approaches. We also consider future research directions as well as the most important barriers to successful clinical translation, emphasizing the transformative potential of tumour-responsive nanomaterials in overcoming the barriers that limit the activity of traditional immunotherapies, thus improving patient outcomes.
Collapse
Affiliation(s)
- Stephen W Linderman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Hospital Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Louis DeRidder
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lucía Sanjurjo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Michael B Foote
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
- IMDEA Nanosciences Institute, Madrid, Spain
| | - Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Robert Langer
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
10
|
Schneider D, Brown EDL, Gluski J, Mishra A, Shah HA, Sciubba DM, Lo SFL. Subtype-Specific Patterns of Tumor Purity and Mutation Load Suggest Treatment Implications: A Cross-Sectional Analysis of 7494 Soft Tissue and Bone Sarcomas (MSK Cohort). Am J Clin Oncol 2025; 48:185-192. [PMID: 40085522 DOI: 10.1097/coc.0000000000001161] [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: 03/16/2025]
Abstract
OBJECTIVES Sarcomas are complex mesenchymal malignancies whose molecular characteristics can significantly influence treatment strategies. This study aimed to investigate the relationship between tumor purity, mutation load, and clinical characteristics across sarcoma subtypes, focusing on potential implications for therapeutic stratification. METHODS This study analyzed the molecular characteristics of 7494 sarcoma cases from the Soft Tissue and Bone Sarcoma (MSK, Nat Commun 2022) data set using available case analysis. Correlations between tumor purity, mutation load, age, and sex were analyzed using nonparametric methods, with subtype-specific analyses conducted using Kruskal-Wallis tests and Bonferroni-corrected post hoc comparisons. A comprehensive analysis of mutation patterns was performed using microsatellite instability (MSI) status. RESULTS Significant correlations between mutation load and tumor purity (ρ=0.320, P <0.001) were identified, with marked heterogeneity across subtypes. Tumor purity ranged from 20.0% in brain sarcomas to 78.5% in dermatofibrosarcoma protuberans. Age-related molecular changes were observed in brain (ρ=0.711, P =0.006) and skin sarcomas (ρ=0.450, P =0.006), suggesting distinct evolutionary patterns. A subset of hypermutated, microsatellite stable cases (0.15%) with mutation loads exceeding 100 mutations/mb were identified, suggesting alternative mechanisms of genomic instability. MSI-high status was rare (0.24%) but associated with higher mutation loads (median: 25.84 vs. 2.42, P <0.001), particularly in uterine sarcomas (0.7% prevalence). CONCLUSIONS The identification of distinct molecular patterns across sarcoma subtypes challenge existing morphology-based classification systems and may hold implications for therapeutic stratification. These findings may help inform future immunotherapeutic and molecular-guided approaches to treatment in sarcoma patients, particularly for elderly patients with brain sarcomas or females with uterine sarcomas.
Collapse
Affiliation(s)
- Daniel Schneider
- Department of Neurosurgery, Donald and Barbara Zucker Hofstra School of Medicine at Northwell, Manhasset, NY
| | | | | | | | | | | | | |
Collapse
|
11
|
Ziogas DC, Theocharopoulos C, Aravantinou K, Boukouris AE, Stefanou D, Anastasopoulou A, Lialios PP, Lyrarakis G, Gogas H. Clinical benefit of immune checkpoint inhibitors in elderly cancer patients: Current evidence from immunosenescence pathophysiology to clinical trial results. Crit Rev Oncol Hematol 2025; 208:104635. [PMID: 39889861 DOI: 10.1016/j.critrevonc.2025.104635] [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: 06/27/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
The age-related decline in immunity appears to be associated not only with cancer development but also with differential responses to immune checkpoint inhibitors (ICIs). Despite their increasing utility across various malignancies and therapeutic settings, limited data -derived primarily from subgroup analyses of randomized controlled trials (RCTs), pooled meta-analyses, and retrospective studies- are available on the effects of aging on their efficacy and toxicity. Immunosenescence, characterized by the progressive decline of the function of the immune system, and inflammaging, a state of persistent low-grade sterile inflammation, may influence ICI outcomes. Additionally, the incidence, severity, and subtypes of immune-related adverse events (irAEs) may differ between older and younger individuals due to loss of immunotolerance. In the current review, starting from a a comprehensive discussion of the pathophysiology of immunosenescence, we proceed to critically review age-related retrospective and randomized evidence supporting FDA-approved ICIs. We highlight similarities or differences across age groups and the clinical benefit of ICIs in elderly versus younger cancer patients. The optimal integration of ICIs in geriatric oncology necessitates greater inclusion of this patient demographic in RCTs along with real-world data in order to acquire robust data which will guide evidence-based treatment decisions for this population.
Collapse
Affiliation(s)
- Dimitrios C Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Charalampos Theocharopoulos
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Katerina Aravantinou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Aristeidis E Boukouris
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Dimitra Stefanou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Amalia Anastasopoulou
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Panagiotis-Petros Lialios
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - George Lyrarakis
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens School of Medicine, Athens, Greece.
| |
Collapse
|
12
|
Eskandari A, Leow TC, Rahman MBA, Oslan SN. Advances in Therapeutic Cancer Vaccines, Their Obstacles, and Prospects Toward Tumor Immunotherapy. Mol Biotechnol 2025; 67:1336-1366. [PMID: 38625508 DOI: 10.1007/s12033-024-01144-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/15/2024] [Indexed: 04/17/2024]
Abstract
Over the past few decades, cancer immunotherapy has experienced a significant revolution due to the advancements in immune checkpoint inhibitors (ICIs) and adoptive cell therapies (ACTs), along with their regulatory approvals. In recent times, there has been hope in the effectiveness of cancer vaccines for therapy as they have been able to stimulate de novo T-cell reactions against tumor antigens. These tumor antigens include both tumor-associated antigen (TAA) and tumor-specific antigen (TSA). Nevertheless, the constant quest to fully achieve these abilities persists. Therefore, this review offers a broad perspective on the existing status of cancer immunizations. Cancer vaccine design has been revolutionized due to the advancements made in antigen selection, the development of antigen delivery systems, and a deeper understanding of the strategic intricacies involved in effective antigen presentation. In addition, this review addresses the present condition of clinical tests and deliberates on their approaches, with a particular emphasis on the immunogenicity specific to tumors and the evaluation of effectiveness against tumors. Nevertheless, the ongoing clinical endeavors to create cancer vaccines have failed to produce remarkable clinical results as a result of substantial obstacles, such as the suppression of the tumor immune microenvironment, the identification of suitable candidates, the assessment of immune responses, and the acceleration of vaccine production. Hence, there are possibilities for the industry to overcome challenges and enhance patient results in the coming years. This can be achieved by recognizing the intricate nature of clinical issues and continuously working toward surpassing existing limitations.
Collapse
Affiliation(s)
- Azadeh Eskandari
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia.
| | - Thean Chor Leow
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| | | | - Siti Nurbaya Oslan
- Enzyme and Microbial Technology Research Centre, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
- Enzyme Technology and X-ray Crystallography Laboratory, VacBio 5, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM, Serdang, Selangor, Malaysia
| |
Collapse
|
13
|
Kamali MJ, Salehi M, Fath MK. Advancing personalized immunotherapy for melanoma: Integrating immunoinformatics in multi-epitope vaccine development, neoantigen identification via NGS, and immune simulation evaluation. Comput Biol Med 2025; 188:109885. [PMID: 40010174 DOI: 10.1016/j.compbiomed.2025.109885] [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: 11/25/2024] [Revised: 01/23/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
The use of cancer vaccines represents a promising avenue in cancer immunotherapy. Advances in next-generation sequencing (NGS) technology, coupled with the development of sophisticated analysis tools, have enabled the identification of somatic mutations by comparing genetic sequences between normal and tumor samples. Tumor neoantigens, derived from these mutations, have emerged as potential candidates for therapeutic cancer vaccines. In this study, raw NGS data from two melanoma patients (NCI_3903 and NCI_3998) were analyzed using publicly available SRA datasets from NCBI to identify patient-specific neoantigens. A comprehensive pipeline was employed to select candidate peptides based on their antigenicity, immunogenicity, physicochemical properties, and toxicity profiles. These validated epitopes were utilized to design multi-epitope chimeric vaccines tailored to each patient. Peptide linkers were employed to connect the epitopes, ensuring optimal vaccine structure and function. The two-dimensional (2D) and three-dimensional (3D) structures of the chimeric vaccines were predicted and refined to ensure structural stability and immunogenicity. Furthermore, molecular docking simulations were conducted to evaluate the binding interactions between the vaccine chimeras and the HLA class I receptors, confirming their potential to elicit a robust immune response. This work highlights a personalized approach to cancer vaccine development, demonstrating the feasibility of utilizing neoantigen-based immunoinformatics pipelines to design patient-specific therapeutic vaccines for melanoma.
Collapse
Affiliation(s)
- Mohammad Javad Kamali
- Department of Medical Genetics, School of Medicine, Babol University of Medical Science, Babol, Iran
| | - Mohammad Salehi
- Department of Medical Genetics, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Science, Kharazmi University, Tehran, Iran.
| |
Collapse
|
14
|
Poudel K, Ji Z, Njauw CN, Rajadurai A, Bhayana B, Sullivan RJ, Kim JO, Tsao H. Fabrication and functional validation of a hybrid biomimetic nanovaccine (HBNV) against Kit K641E -mutant melanoma. Bioact Mater 2025; 46:347-364. [PMID: 39834347 PMCID: PMC11742834 DOI: 10.1016/j.bioactmat.2024.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/03/2024] [Accepted: 12/20/2024] [Indexed: 01/22/2025] Open
Abstract
Cancer nanovaccines hold the promise for personalization, precision, and pliability by integrating all the elements essential for effective immune stimulation. An effective immune response requires communication and interplay between antigen-presenting cells (APCs), tumor cells, and immune cells to stimulate, extend, and differentiate antigen-specific and non-specific anti-tumor immune cells. The versatility of nanomedicine can be adapted to deliver both immunoadjuvant payloads and antigens from the key players in immunity (i.e., APCs and tumor cells). The imperative for novel cancer medicine is particularly pressing for less common but more devastating KIT-mutated acral and mucosal melanomas that are resistant to small molecule c-kit and immune checkpoint inhibitors. To overcome this challenge, we successfully engineered nanotechnology-enabled hybrid biomimetic nanovaccine (HBNV) comprised of membrane proteins (antigens to activate immunity and homing/targeting ligand to tumor microenvironment (TME) and lymphoid organs) from fused cells (of APCs and tumor cells) and immunoadjuvant. These HBNVs are efficiently internalized to the target cells, assisted in the maturation of APCs via antigens and adjuvant, activated the release of anti-tumor cytokines/inhibited the release of immunosuppressive cytokine, showed a homotypic effect on TME and lymph nodes, activated the anti-tumor immune cells/downregulated the immunosuppressive immune cells, reprogram the tumor microenvironment, and showed successful anti-tumor therapeutic and prophylactic effects.
Collapse
Affiliation(s)
- Kishwor Poudel
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhenyu Ji
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ching-Ni Njauw
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anpuchchelvi Rajadurai
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Brijesh Bhayana
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryan J. Sullivan
- Mass General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Hensin Tsao
- Wellman Center for Photomedicine and Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Mass General Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Guo X, Bai J, Wang X, Guo S, Shang Z, Shao Z. Evoking the Cancer-immunity cycle by targeting the tumor-specific antigens in Cancer immunotherapy. Int Immunopharmacol 2025; 154:114576. [PMID: 40168803 DOI: 10.1016/j.intimp.2025.114576] [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: 02/08/2025] [Revised: 03/17/2025] [Accepted: 03/27/2025] [Indexed: 04/03/2025]
Abstract
Cancer-related deaths continue to rise, largely due to the suboptimal efficacy of current treatments. Fortunately, immunotherapy has emerged as a promising alternative, offering new hope for cancer patients. Among various immunotherapy approaches, targeting tumor-specific antigens (TSAs) has gained particular attention due to its demonstrated success in clinical settings. Despite these advancements, there are still gaps in our understanding of TSAs. Therefore, this review explores the life cycle of TSAs in cancer, the methods used to identify them, and recent advances in TSAs-targeted cancer therapies. Enhancing medical professionals' understanding of TSAs will help facilitate the development of more effective TSAs-based cancer treatments.
Collapse
Affiliation(s)
- Xiaomeng Guo
- 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
| | - Junqiang Bai
- 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
| | - Xinmiao 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
| | - Shutian Guo
- 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
| | - Zhengjun Shang
- 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-Head and Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Zhe Shao
- 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; Day Surgery Center, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
16
|
Alenezi SK. CAR T cells in lung cancer: Targeting tumor-associated antigens to revolutionize immunotherapy. Pathol Res Pract 2025; 269:155947. [PMID: 40168775 DOI: 10.1016/j.prp.2025.155947] [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: 01/18/2025] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
Tumor-targeted T cells engineered for targeting and killing tumor cells have revolutionized cancer treatment, specifically in hematologic malignancies, through chimeric antigen receptor (CAR) T cell therapy. However, the migration of this success to lung cancer is challenging due to the tumor microenvironment (TME), antigen heterogeneity, and limitations of T cell infiltration. This review aims to evaluate current strategies addressing these barriers, focusing on the optimization of tumor-associated antigen (TAA) targeting, such as epidermal growth factor receptor (EGFR), mucin-1 (MUC1), and mesothelin (MSLN), which are frequently overexpressed in lung cancer and offer promising targets for CAR T-cell therapy. In this review, we discuss recent progress in CAR T cell engineering, applying enhanced costimulatory molecules, cytokine-secreting CAR T cells, and engineered modifications to improve T cell resilience in immunosuppressive environments. Additionally, this review also evaluates combination therapies of immune checkpoint inhibitors and recently published clinical trials on lung cancer with CAR T cells. We offer insights into the way to optimize CAR T cell therapy for lung cancer by analyzing antigen selection, immune evasion, and the strategies to enhance T cell persistence and tumor infiltration.
Collapse
Affiliation(s)
- Sattam Khulaif Alenezi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al-Qassim 51452, Saudi Arabia.
| |
Collapse
|
17
|
Mivehchi H, Eskandari-Yaghbastlo A, Ghazanfarpour M, Ziaei S, Mesgari H, Faghihinia F, Zokaei Ashtiani N, Afjadi MN. Microenvironment-based immunotherapy in oral cancer: a comprehensive review. Med Oncol 2025; 42:140. [PMID: 40153139 DOI: 10.1007/s12032-025-02694-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/19/2025] [Indexed: 03/30/2025]
Abstract
Oral cancer, a prevalent form of head and neck malignancy, accounts for 4% of global cancer cases. The most common type, oral squamous cell carcinoma (OSCC), has a survival rate of about 50%. Even though emerging molecular therapies show promise for managing oral cancer, current treatments like surgery, radiotherapy, and chemotherapy have significant side effects. In addition, the complex tumor microenvironment (TME), involving the extracellular matrix (ECM) and cells like fibroblasts and stromal cells like immune cells, promotes tumor growth and inhibits immune responses, complicating treatment. Nonetheless, immunotherapy is crucial in cancer treatment, especially in oral cancers. Indeed, its effectiveness lies in targeting immune checkpoints such as PD-1 and CTLA-4 inhibitors, as well as monoclonal antibodies like pembrolizumab and cetuximab, adoptive cell transfer methods (including CAR-T cell therapy), cytokine therapy such as IL-2, and tumor vaccines. Thus, these interventions collectively regulate tumor proliferation and metastasis by targeting the TME through autocrine-paracrine signaling pathways. Immunotherapy indeed aims to stimulate the immune system, leveraging both innate and adaptive immunity to counteract cancer cell signals and promote tumor destruction. This review will explore how the TME controls tumor proliferation and metastasis via autocrine-paracrine signaling pathways. It will then detail the effectiveness of immunotherapy in oral cancers, focusing on immune checkpoints, targeted monoclonal antibodies, adoptive cell transfer, cytokine therapy, and tumor vaccines.
Collapse
Affiliation(s)
- Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | | | - SeyedMehdi Ziaei
- Faculty of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hassan Mesgari
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Islamic Azad University, Tehran Branch, Tehran, Iran
| | - Farbod Faghihinia
- School of Dentistry, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
18
|
Zheng Y, Wang B, Cai Z, Lai Z, Yu H, Wu M, Liu X, Zhang D. Tailoring nanovectors for optimal neoantigen vaccine efficacy. J Mater Chem B 2025; 13:4045-4058. [PMID: 40042164 DOI: 10.1039/d4tb02547d] [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: 03/27/2025]
Abstract
The primary objective of neoantigen vaccines is to elicit a robust anti-tumor immune response by generating neoantigen-specific T cells that can eradicate tumor cells. Despite substantial advancements in personalized neoantigen prediction using next-generation sequencing, machine learning, and mass spectrometry, challenges remain in efficiently expanding neoantigen-specific T cell populations in vivo. This challenge impedes the widespread clinical application of neoantigen vaccines. Nanovector-based neoantigen delivery systems have emerged as a promising solutions to this challenge. These nanovectors offer several advantages, such as enhanced stability, targeted intracellular delivery, sustained release, and improved antigen-presenting cell (APC) activation. Notably, they effectively deliver various neoantigen vaccine formulations (DC cell-based, synthetic long peptide (SLP)-based or DNA/mRNA-based) to APCs or T cells, thereby activating both CD4+ T and CD8+ T cells. This ultimately induces a specific anti-tumor immune response. This review focuses on recent innovations in neoantigen vaccine delivery vectors. We aim to identify optimal design parameters for vectors tailored to different neoantigen vaccine types, with an emphasis on enhancing the tumor microenvironment and stimulating the production of neoantigen-specific cytotoxic T cells. By maximizing the potential of these delivery systems, we aim to accelerate the clinical translation of neoantigen nanovaccines and advance cancer immunotherapy.
Collapse
Affiliation(s)
- Youshi Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Bing Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Zisen Lai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
| | - Haijun Yu
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China.
- Fujian Agriculture and Forestry University, Fuzhou 350002, P. R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| |
Collapse
|
19
|
Tiwade PB, Fung V, VanKeulen-Miller R, Narasipura EA, Ma Y, Fenton OS. Non-Viral RNA Therapies for Non-Small Cell Lung Cancer and Their Corresponding Clinical Trials. Mol Pharm 2025. [PMID: 40131145 DOI: 10.1021/acs.molpharmaceut.4c00871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Ribonucleic acid (RNA)-based therapies represent a promising class of drugs for the treatment of non-small cell lung cancer (NSCLC) due to their ability to modulate gene expression. Therapies leveraging small interfering RNA (siRNA), messenger RNA (mRNA), microRNA (miRNA), and antisense oligonucleotides (ASOs) offer various advantages over conventional treatments, including the ability to target specific genetic mutations and the potential for personalized medicine approaches. However, the clinical translation of these therapeutics for the treatment of NSCLC faces challenges in delivery due to their immunogenicity, negative charge, and large size, which can be mitigated with delivery platforms. In this review, we provide a description of the pathophysiology of NSCLC and an overview of RNA-based therapeutics, specifically highlighting their potential application in the treatment of NSCLC. We discuss relevant classes of RNA and their therapeutic potential for NSCLC. We then discuss challenges in delivery and non-viral delivery strategies such as lipid- and polymer-based nanoparticles that have been developed to address these issues in preclinical models. Furthermore, we provide a summary table of clinical trials that leverage RNA therapies for NSCLC [which includes their National Clinical Trial (NCT) numbers] to highlight the current progress in NSCLC. We also discuss how these NSCLC therapies can be integrated with existing treatment modalities to enhance their efficacy and improve patient outcomes. Overall, we aim to highlight non-viral strategies that tackle RNA delivery challenges while showcasing RNA's potential as a next-generation therapy for NSCLC treatment.
Collapse
Affiliation(s)
- Palas Balakdas Tiwade
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Vincent Fung
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachel VanKeulen-Miller
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Eshan Amruth Narasipura
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yutian Ma
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Owen S Fenton
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
20
|
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] [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
|
21
|
Narote S, Desai SA, Patel VP, Deshmukh R, Raut N, Dapse S. Identification of new immune target and signaling for cancer immunotherapy. Cancer Genet 2025; 294-295:57-75. [PMID: 40154216 DOI: 10.1016/j.cancergen.2025.03.004] [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: 01/01/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Immunotherapy has become one of the innovative treatments in malignancy as it activates the immune system to find and eliminate malignant cells. The tumor immunology interface has become increasingly intricate, making the identification of new immune targets and signalling pathways on which to base improved therapeutic strategies an ongoing process. This review, we goal to clarify the contacts between cancer and immune system with a focus on immune surveillance as well as immune evasion mechanisms. Comprehensive immunotherapeutic therapies are overviewed with ICI (CTLA-4, PD-1, PD-L1), CAR-T cell therapy, and cancer vaccines whereas, advanced therapies targeting new immune checkpoints are also elucidated including TIM-3, LAG-3, and TIGIT. The JAK/STAT, MAPK and PI3K-AKT-mTOR pathways are reviewed with regards to cancer progression and immunotherapeutic resistance. The dysregulation of these pathways gives hope for the identification of fresh targets for therapy. Genomics, proteomics, immunopeptidomics, single cell mass spectrometry, CRISPR-based functional genomics and bioinformatics are described as essential for immune target identification and for mapping of cancer relevant signaling pathways. This review also considers some emerging issues in the subject area like the tumor heterogeneity, immune-related adverse events (irAEs), and personalized treatment. These barriers are described to facilitate the understanding of ways to overcome them and increase the efficacy of immunotherapies through combination therapies. This means that by developing new knowledge of immunological targets and pathways, immunoprecision medicine for cancer could greatly enhance outcomes.
Collapse
Affiliation(s)
- Sakshi Narote
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sharav A Desai
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India.
| | - Vipul P Patel
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Rutuja Deshmukh
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Nikita Raut
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sejal Dapse
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| |
Collapse
|
22
|
Wu M, Liu J, Liu L, Yang Y, Liu H, Yu L, Zeng H, Yuan S, Xu R, Liu H, Jiang H, Qu S, Wang L, Chen Y, Wang J, Zhang Y, He S, Feng L, Han J, Zeng W, Wang H, Huang Y. Autologous Peripheral Vγ9Vδ2 T Cell Synergizes with αβ T Cell Through Antigen Presentation and BTN3A1 Blockade in Immunotherapy of Cervical Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2401230. [PMID: 40091603 DOI: 10.1002/advs.202401230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 02/07/2025] [Indexed: 03/19/2025]
Abstract
New treatment strategies are urgently needed for patients with advanced cervical cancer (CC). Here, a synergistic anti-CC effect of a novel combinatorial immunotherapy with adoptively transferred autologous Vγ9Vδ2 T cells and αβ T cells is shown. The pivotal role of both circulating and tumor-infiltrating Vγ9Vδ2 T cells in anti-CC immunity is uncovered. Importantly, autologous Vγ9Vδ2 T cells show a synergistic anti-CC effect with αβ T cells not only through killing tumor directly, but also by promoting the activation and tumoricidal activity of syngeneic αβ T cells through antigen presentation, which can be further boosted by conventional chemotherapy. Moreover, Vγ9Vδ2 T cells can restore the tumoricidal function of αβ T cell through competitively binding to BTN3A1, a TCR-Vγ9Vδ2 ligand on CC cells upregulated by IFN-γ derived from activated αβ T cell. These findings uncover a critical synergistic effect of autologous Vγ9Vδ2 T cells and αβ T cells in immunotherapy of CC and reveal the underlying mechanisms.
Collapse
Affiliation(s)
- Min Wu
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Jian Liu
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Liting Liu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Yifan Yang
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hong Liu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Long Yu
- Beckman Coulter Commercial Enterprise (China) Co., Ltd, Shanghai, 200122, China
| | - Haihong Zeng
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shuo Yuan
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Ruiyi Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Hangyu Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Han Jiang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shen Qu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Liming Wang
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingyu Wang
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuwei Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shan He
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ling Feng
- Department of Obstetrics and Gynecology, Tongji Hospital and School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Junyan Han
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wanjiang Zeng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
23
|
Zavadil F, Henek T, Habault J, Chemali R, Tovar-Fernandez MC, Daskalogianni C, Malbert-Colas L, Wang L, Gnanasundram SV, Vojtesek B, Hernychova L, Apcher S, Fahraeus R. Translation of bi-directional transcripts enhances MHC-I peptide diversity. Front Immunol 2025; 16:1554561. [PMID: 40165968 PMCID: PMC11956742 DOI: 10.3389/fimmu.2025.1554561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Antisense transcripts play an important role in generating regulatory non-coding RNAs but whether these transcripts are also translated to generate functional peptides remains poorly understood. In this study, RNA sequencing and six-frame database generation were combined with mass spectrometry analysis of peptides isolated from polysomes to identify Nascent Pioneer Translation Products (Na-PTPs) originating from alternative reading frames of bi-directional transcripts. Two Na-PTP originating peptides derived from antisense strands stimulated CD8+ T cell proliferation when presented to peripheral blood mononuclear cells (PBMCs) from nine healthy donors. Importantly, an antigenic peptide derived from the reverse strand of two cDNA constructs was presented on MHC-I molecules and induced CD8+ T cell activation. The results demonstrate that three-frame translation of bi-directional transcripts generates antigenic peptide substrates for the immune system. This discovery holds significance for understanding the origin of self-discriminating peptide substrates for the major histocompatibility class I (MHC-I) pathway and for enhancing immune-based therapies against infected or transformed cells.
Collapse
Affiliation(s)
- Filip Zavadil
- RECAMO, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Tomas Henek
- RECAMO, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Justine Habault
- Inserm UMRS1131, Institut de Génétique Moléculaire, Université Paris Cité, Hôpital St. Louis, Paris, France
| | - René Chemali
- UMR 1015 Immunologie des tumeurs et immunothérapie contre le cancer, B2M, Gustave Roussy, Université Paris Sud, Villejuif, France
| | | | - Chrysoula Daskalogianni
- Inserm UMRS1131, Institut de Génétique Moléculaire, Université Paris Cité, Hôpital St. Louis, Paris, France
| | - Laurence Malbert-Colas
- Inserm UMRS1131, Institut de Génétique Moléculaire, Université Paris Cité, Hôpital St. Louis, Paris, France
| | - Lixiao Wang
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | | | - Borek Vojtesek
- RECAMO, Masaryk Memorial Cancer Institute, Brno, Czechia
- Laboratory of Growth Regulators, Institute of Experimental Botany, The Czech Academy of Sciences, Olomouc, Czechia
| | | | - Sebastien Apcher
- UMR 1015 Immunologie des tumeurs et immunothérapie contre le cancer, B2M, Gustave Roussy, Université Paris Sud, Villejuif, France
| | - Robin Fahraeus
- RECAMO, Masaryk Memorial Cancer Institute, Brno, Czechia
- Inserm UMRS1131, Institut de Génétique Moléculaire, Université Paris Cité, Hôpital St. Louis, Paris, France
- UMR 1015 Immunologie des tumeurs et immunothérapie contre le cancer, B2M, Gustave Roussy, Université Paris Sud, Villejuif, France
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
- Laboratory of Growth Regulators, Institute of Experimental Botany, The Czech Academy of Sciences, Olomouc, Czechia
| |
Collapse
|
24
|
Takahashi H, Hanaoka K, Wada H, Kojima D, Watanabe M. The Current Status of T Cell Receptor (TCR) Repertoire Analysis in Colorectal Cancer. Int J Mol Sci 2025; 26:2698. [PMID: 40141338 PMCID: PMC11943327 DOI: 10.3390/ijms26062698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/28/2025] Open
Abstract
The rapid increase in colorectal cancer (CRC) cases recently has highlighted the need to use predictive biomarkers to guide therapeutic approaches. Current studies have focused on the tumor-infiltrating lymphocytes present in the tumor microenvironment (TME), in which cytotoxic T cell activation and the amount are associated with CRC patient prognosis. The T cell receptor (TCR) is essential for antigen recognition and T cell identification, playing a central role in cancer immunotherapy. The T cell status reflects TCR diversity or clonality, known as the TCR repertoire. Accordingly, analyzing the TCR repertoire dynamics may help predict the immunological circumstances of the TME in a timely way. In this review, we summarize the TCR repertoire-related knowledge, including its potential use as predictive biomarkers in CRC. The intratumoral TCR repertoire is restricted in CRC patients compared with healthy individuals, as well as in peripheral blood. Patients with deficient mismatch repair display more restriction than those with proficient mismatch repair. Importantly, a higher TCR diversity before treatment and a decrease following treatment may indicate a good response and a better clinical outcome in CRC patients. The future use of TCR repertoire sequencing technology combined with artificial intelligence-based analysis is a potential strategy for CRC therapeutic decision making.
Collapse
Affiliation(s)
- Hiroyuki Takahashi
- Department of Surgery, Fukuoka University Chikushi Hospital, 1-1-1 Zokumyoin, Chikushino 818-8502, Fukuoka, Japan; (K.H.); (H.W.); (D.K.); (M.W.)
| | | | | | | | | |
Collapse
|
25
|
Arnet L, Emilius L, Hamann A, Carmo-Fonseca M, Berking C, Dörrie J, Schaft N. The Influence of Indisulam on Human Immune Effector Cells: Is a Combination with Immunotherapy Feasible? Pharmaceutics 2025; 17:368. [PMID: 40143032 PMCID: PMC11945250 DOI: 10.3390/pharmaceutics17030368] [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: 07/31/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Background: As a modulator of pre-mRNA splicing, the anti-cancer agent indisulam can induce aberrantly spliced neoantigens, enabling immunologic anti-tumor activity. Consequently, combining indisulam with immunotherapy is expected to be a promising novel approach in cancer therapy. However, a prerequisite for such a combination is that immune effector cells remain functional and unharmed by the chemical. Methods: To ensure the immunocompetence of human immune effector cells is maintained, we investigated the influence of indisulam on ex vivo-isolated T cells and monocyte-derived dendritic cells (moDCs) from healthy donors. We used indisulam concentrations from 0.625 µM to 160 µM and examined the impact on the following: (i) the activation of CD4+ and CD8+ T cells by CD3-crosslinking and via a high-affinity TCR, (ii) the cytotoxicity of CD8+ T cells, (iii) the maturation process of moDCs, and (iv) antigen-specific CD8+ T cell priming. Results: We observed dose-dependent inhibitory effects of indisulam, and substantial inhibition occurred at concentrations around 10 µM, but the various functions of the immune system exhibited different sensitivities. The weaker activation of T cells via CD3-crosslinking was more sensitive than the stronger activation via the high-affinity TCR. T cells remained capable of killing tumor cells after treatment with indisulam up to 40 µM, but T cell cytotoxicity was impaired at 160 µM indisulam. While moDC maturation was also rather resistant, T cell priming was almost completely abolished at a concentration of 10 µM. Conclusions: These effects should be considered in possible future combinations of immunotherapy with the mRNA splicing inhibitor indisulam.
Collapse
Affiliation(s)
- Lisa Arnet
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Lisabeth Emilius
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Annett Hamann
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Maria Carmo-Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| |
Collapse
|
26
|
Singh P, Khatib MN, R R, Kaur M, Srivastava M, Barwal A, Rajput GVS, Rajput P, Syed R, Sharma G, Kumar S, Shabil M, Pandey S, Brar M, Bushi G, Mehta R, Sah S, Goh KW, Satapathy P, Gaidhane AM, Samal SK. Advancements and challenges in personalized neoantigen-based cancer vaccines. Oncol Rev 2025; 19:1541326. [PMID: 40160263 PMCID: PMC11949952 DOI: 10.3389/or.2025.1541326] [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: 12/07/2024] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
Advancements in personalized neoantigen-based cancer vaccines are ushering in a new era in oncology, targeting unique genetic alterations within tumors to enhance treatment precision and efficacy. Neoantigens, specific to cancer cells and absent in normal tissues, are at the heart of these vaccines, promising to direct the immune system specifically against the tumor, thereby maximizing therapeutic efficacy while minimizing side effects. The identification of neoantigens through genomic and proteomic technologies is central to developing these vaccines, allowing for the precise mapping of a tumor's mutational landscape. Despite advancements, accurately predicting which neoantigens will elicit strong immune responses remains challenging due to tumor variability and the complexity of immune system interactions. This necessitates further refinement of bioinformatics tools and predictive models. Moreover, the efficacy of these vaccines heavily depends on innovative delivery methods that enhance neoantigen presentation to the immune system. Techniques like encapsulating neoantigens in lipid nanoparticles and using viral vectors are critical for improving vaccine stability and delivery. Additionally, these vaccines contribute towards achieving Sustainable Development Goal 3.8, promoting universal health coverage by advancing access to safe and effective cancer treatments. This review delves into the potential of neoantigen-based vaccines to transform cancer treatment, examining both revolutionary advancements and the ongoing challenges they face.
Collapse
Affiliation(s)
- Parminder Singh
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia
| | - Mahalaqua Nazli Khatib
- Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education, Wardha, India
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mandeep Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | | | - Amit Barwal
- Chandigarh Pharmacy College, Chandigarh Group of College, Mohali, Punjab, India
| | - G. V. Siva Rajput
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Pranchal Rajput
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Rukshar Syed
- IES Institute of Pharmacy, IES University, Bhopal, Madhya Pradesh, India
| | - Gajendra Sharma
- New Delhi Institute of Management, Tughlakabad Institutional Area, New Delhi, India
| | - Sunil Kumar
- Department of Microbiology, Graphic Era (Deemed to be University), Dehradun, India
| | - Muhammed Shabil
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Sakshi Pandey
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India
| | - Manvinder Brar
- Chitkara Centre for Research and Development, Chitkara University, Solan, Himachal Pradesh, India
| | - Ganesh Bushi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Rachana Mehta
- Clinical Microbiology, RDC, Manav Rachna International Institute of Research and Studies, Faridabad, Haryana, India
| | - Sanjit Sah
- Department of Paediatrics, Dr. D. Y. Patil Medical College Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed-to-be-University), Pimpri, Pune, Maharashtra, India
- Department of Public Health Dentistry, Dr. D. Y. Patil Medical College Hospital and Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed-to-be-University), Pimpri, Pune, Maharashtra, India
- Department of Medicine, Korea Universtiy, Seoul, Republic of Korea
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia
- Faculty of Mathematics and Natural Sciences, Universitas Negeri Padang, Padang, Indonesia
| | - Prakasini Satapathy
- University Center for Research and Development, Chandigarh University, Mohali, Punjab, India
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Abhay M. Gaidhane
- Jawaharlal Nehru Medical College, and Global Health Academy, School of Epidemiology and Public Health, Datta Meghe Institute of Higher Education, Wardha, India
| | - Shailesh Kumar Samal
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
27
|
Coelho MO, Quintas ST, Sarmento B, De Wever O, Castro F. Engineered dendritic cells-derived extracellular vesicles for cancer immunotherapy. J Control Release 2025; 381:113620. [PMID: 40088976 DOI: 10.1016/j.jconrel.2025.113620] [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: 12/10/2024] [Revised: 03/03/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
Extracellular vesicles (EVs) have emerged as a cell-free therapeutic approach, garnering increasing attention for their potential to enhance the safety and efficacy of immunotherapy. This interest is primarily driven by the biocompatibility and cell/tissue tropism inherent to EVs, but also due to their reconfigurable content. This, termed as cargo, may comprise bioactive molecules as proteins, lipids, and nucleic acids that play a pivotal role in mediating intercellular communication. In particular, dendritic cells-derived extracellular vesicles (DC-EVs) facilitate the transfer of critical components, like antigens and immune-regulatory factors, and due to the expression of major histocompatibility complexes and co-stimulatory molecules on their surface can activate T cells, thereby modulating the immune response. Additionally, DC-EVs can be engineered to transport tumor-specific antigens, cytokines, or other agents in order to strength their immunotherapeutic potential, and even be used in vaccines formulation. In this review, the latest advancements in engineering DC-EVs to improve their immunotherapeutic potential is discussed in detail, while also addressing current challenges associated with DC-EVs therapies.
Collapse
Affiliation(s)
- Margarida Oliveira Coelho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Sofia Torres Quintas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal; IUCS-CESPU, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Olivier De Wever
- CRIG - Cancer Research Institute Ghent, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium; LECR - Laboratory Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto 4200-135, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200- 180 Porto, Portugal.
| |
Collapse
|
28
|
Brlek P, Bulić L, Shah N, Shah P, Primorac D. In Silico Validation of OncoOrigin: An Integrative AI Tool for Primary Cancer Site Prediction with Graphical User Interface to Facilitate Clinical Application. Int J Mol Sci 2025; 26:2568. [PMID: 40141210 PMCID: PMC11942019 DOI: 10.3390/ijms26062568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/08/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Cancers of unknown primary (CUPs) represent a significant diagnostic and therapeutic challenge in the field of oncology. Due to the limitations of current diagnostic tools in these cases, novel approaches must be brought forward to improve treatment outcomes for these patients. The objective of this study was to develop a machine-learning-based software for primary cancer site prediction (OncoOrigin), based on genetic data acquired from tumor DNA sequencing. By design, this was an in silico diagnostic study, conducted using data from the cBioPortal database (accessed on 21 September 2024) and several data processing and machine learning Python libraries. The study involved over 20,000 tumor samples with information on patient age, sex, and the presence of genetic variants in over 600 genes. The main outcome of interest was machine-learning-based discrimination between cancer site classes. Model quality was assessed by training set cross-validation and evaluation on a segregated test set. Finally, the optimal model was incorporated with a graphical user interface into the OncoOrigin software. Feature importance for class discrimination was also determined on the optimal model. Out of the four tested machine learning estimators, the XGBoostClassifier-based model proved superior in test set evaluation, with a top-2 accuracy of 0.91 and ROC-AUC of 0.97. Unlike other machine learning models published in the literature, OncoOrigin stands out as the only one integrated with a graphical user interface, which is crucial for facilitating its use by oncology specialists in everyday clinical practice, where its application and implementation will have the greatest value in the future.
Collapse
Affiliation(s)
- Petar Brlek
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (L.B.)
- School of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Luka Bulić
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (L.B.)
| | - Nidhi Shah
- Dartmouth Health, Lebanon, NH 03766, USA
| | - Parth Shah
- Dartmouth Health, Lebanon, NH 03766, USA
| | - Dragan Primorac
- St. Catherine Specialty Hospital, 10000 Zagreb, Croatia; (L.B.)
- School of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Eberly College of Science, The Pennsylvania State University, State College, PA 16802, USA
- School of Medicine, University of Split, 21000 Split, Croatia
- The Henry C. Lee College of Criminal Justice and Forensic Sciences, University of New Haven, New Haven, CT 06516, USA
- Regiomed Kliniken, 96450 Coburg, Germany
- School of Medicine, University of Rijeka, 51000 Rijeka, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- School of Medicine, University of Mostar, 88000 Mostar, Bosnia and Herzegovina
- National Forensic Sciences University, Gandhinagar 382007, India
| |
Collapse
|
29
|
Nguyen CM, Vu TT, Nguyen MN, Tran-Nguyen TS, Huynh CT, Ha QT, Nguyen HN, Tran LS. Neoantigen-based mRNA vaccine exhibits superior anti-tumor activity compared to synthetic long peptides in an in vivo lung carcinoma model. Cancer Immunol Immunother 2025; 74:145. [PMID: 40072566 PMCID: PMC11949242 DOI: 10.1007/s00262-025-03992-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025]
Abstract
Neoantigen vaccines hold great promise in cancer immunotherapy, but the comparative efficacy of different vaccine platforms, particularly in the context of tumor burden (TB), remains insufficiently studied. In this research, we evaluated the safety and therapeutic efficacy of synthetic long peptide and mRNA-based vaccines, both designed to target identical neoantigens across different Lewis Lung Carcinoma (LLC) tumor burdens. We employed the LLC syngeneic mouse model, a widely used preclinical model for aggressive and immunosuppressive tumors. Our findings demonstrated that the mRNA-based vaccine significantly outperformed the peptide-based vaccine in preventing tumor growth in mice with low TB. These results underscore the potential of mRNA vaccines as a more effective approach for treating aggressive tumors, contributing valuable insights for the future development of neoantigen-based cancer vaccines.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Mice
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/therapy
- mRNA Vaccines/immunology
- Lung Neoplasms/immunology
- Lung Neoplasms/therapy
- Mice, Inbred C57BL
- Disease Models, Animal
- Peptides/immunology
- Immunotherapy/methods
- Female
- Humans
- Vaccines, Synthetic/immunology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/therapeutic use
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- Cell Line, Tumor
Collapse
Affiliation(s)
| | - Trung T Vu
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| | | | | | - Chi Thien Huynh
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Quang Thanh Ha
- Biotechnology Center of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | | | - Le Son Tran
- Medical Genetics Institute, Ho Chi Minh City, Vietnam.
| |
Collapse
|
30
|
Che J, Liu Y, Liu Y, Song J, Cui H, Feng D, Tian A, Zhang Z, Xu Y. The application of emerging immunotherapy in the treatment of prostate cancer: progress, dilemma and promise. Front Immunol 2025; 16:1544882. [PMID: 40145100 PMCID: PMC11937122 DOI: 10.3389/fimmu.2025.1544882] [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: 12/13/2024] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
In recent years, there has been a growing trend towards the utilization of immunotherapy techniques for the treatment of cancer. Some malignancies have acquired significant progress with the use of cancer vaccines, immune checkpoint inhibitors, and adoptive cells therapy. Scholars are exploring the aforementioned methods as potential treatments for advanced prostate cancer (PCa) due to the absence of effective adjuvant therapy to improve the prognosis of metastatic castration-resistant prostate cancer (mCRPC). Immunotherapy strategies have yet to achieve significant advancements in the treatment of PCa, largely attributed to the inhibitory tumor microenvironment and low mutation load characteristic of this malignancy. Hence, researchers endeavor to address these challenges by optimizing the design and efficacy of immunotherapy approaches, as well as integrating them with other therapeutic modalities. To date, studies have also shown potential clinical benefits. This comprehensive review analyzed the utilization of immunotherapy techniques in the treatment of PCa, assessing their advantages and obstacles, with the aim of providing healthcare professionals and scholars with a comprehensive understanding of the progress in this field.
Collapse
Affiliation(s)
- Jizhong Che
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yuanyuan Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yangyang Liu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Jingheng Song
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Hongguo Cui
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Dongdong Feng
- Department of Urology, Haiyang City People’s Hospital, Yantai, Shandong, China
| | - Aimin Tian
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Zhengchao Zhang
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Yankai Xu
- Department of Urology, Yantai Affiliated Hospital of Binzhou Medical University, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
31
|
He W, Cui K, Farooq MA, Huang N, Zhu S, Jiang D, Zhang X, Chen J, Liu Y, Xu G. TCR-T cell therapy for solid tumors: challenges and emerging solutions. Front Pharmacol 2025; 16:1493346. [PMID: 40129944 PMCID: PMC11931055 DOI: 10.3389/fphar.2025.1493346] [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: 09/09/2024] [Accepted: 02/20/2025] [Indexed: 03/26/2025] Open
Abstract
With the use of T cell receptor T cells (TCR-T cells) and chimeric antigen receptor T cells (CAR-T cells), T-cell immunotherapy for cancer has advanced significantly in recent years. CAR-T cell therapy has demonstrated extraordinary success when used to treat hematologic malignancies. Nevertheless, there are several barriers that prevent this achievement from being applied to solid tumors, such as challenges with tumor targeting and inadequate transit and adaption of genetically modified T-cells, especially in unfavorable tumor microenvironments The deficiencies of CAR-T cell therapy in the treatment of solid tumors are compensated for by TCR-T cells, which have a stronger homing ability to initiate intracellular commands, 90% of the proteins can be used as developmental targets, and they can recognize target antigens more broadly. As a result, TCR-T cells may be more effective in treating solid tumors. In this review, we discussed the structure of TCR-T and have outlined the drawbacks of TCR-T in cancer therapy, and suggested potential remedies. This review is crucial in understanding the current state and future potential of TCR-T cell therapy. We emphasize how important it is to use combinatorial approaches, combining new combinations of various emerging strategies with over-the-counter therapies designed for TCR-T, to increase the anti-tumor efficacy of TCR-T inside the TME and maximize treatment safety, especially when it comes to solid tumor immunotherapies.
Collapse
Affiliation(s)
- Wanjun He
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| | - Kai Cui
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| | - Muhammad Asad Farooq
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Na Huang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| | - Songshan Zhu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| | - Dan Jiang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| | - Xiqian Zhang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
- Yinchuan Guolong Orthopedic Hospital, Yinchuan, China
| | - Jian Chen
- Yinchuan Guolong Orthopedic Hospital, Yinchuan, China
| | - Yinxia Liu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Guangxian Xu
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, The First Dongguan Affiliated Hospital, School of Medical Technology, Guangdong Medical University, Dongguan, China
- Dongguan Key Laboratory of Molecular Immunology and Cell Therapy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
32
|
Kim D, Choi J, Jin D, Xu E, Lee J, Byun J, Oh YK. Hybrid lipid nanoparticles with tumor antigen-primed dendritic cell membranes for post-surgical tumor immunotherapy. J Control Release 2025; 379:537-548. [PMID: 39800239 DOI: 10.1016/j.jconrel.2025.01.027] [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: 09/15/2024] [Revised: 12/22/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Post-surgical tumor recurrence poses a major challenge in cancer treatment due to residual tumor cells and surgery-induced immunosuppression. Here, we developed hybrid nanoparticles, termed T-DCNPs, designed to promote antigen-specific activation of cytotoxic CD8+ T cells while concurrently inhibiting immunosuppressive pathways within the tumor microenvironment. T-DCNPs were formulated by co-extruding lipid nanoparticles containing a transforming growth factor β inhibitor with dendritic cells that were pre-treated with autologous neoantigens derived from surgically excised tumors. By using whole tumor antigens rather than specific peptides, T-DCNPs effectively overcame tumor heterogeneity and elicited a robust, targeted immune response. In vitro studies showed that T-DCNPs enhanced CD8+ T cell proliferation and reduced programmed death-1 (PD-1) expression, leading to increased antitumor cytotoxicity. In vivo experiments, involving intratumoral injections of T-DCNPs in distant tumor and post-surgical melanoma models, demonstrated a significant reduction in distant tumor growth, decreased recurrence rates, and extended survival compared to control groups. Flow cytometry and immunohistochemistry analyses further confirmed the enhanced infiltration of activated CD8+ T cells and a marked reduction in immunosuppressive markers, including PD-1 and Foxp3, within the treated tumors. These results suggest that T-DCNPs, through the dual mechanisms of tumor antigen-specific T cell activation and immune modulation, offer a promising strategy to prevent tumor recurrence following surgery and could potentially improve the efficacy of postoperative cancer immunotherapy.
Collapse
Affiliation(s)
- Dongyoon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaehyun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Dongun Jin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Enzhen Xu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea.
| | - Junho Byun
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
33
|
Bedeschi M, Cavassi E, Romeo A, Tesei A. Glioblastoma Tumor Microenvironment and Purinergic Signaling: Implications for Novel Therapies. Pharmaceuticals (Basel) 2025; 18:385. [PMID: 40143161 PMCID: PMC11944773 DOI: 10.3390/ph18030385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Glial-origin brain tumors, particularly glioblastomas (GBMs), are known for their devastating prognosis and are characterized by rapid progression and fatal outcomes. Despite advances in surgical resection, complete removal of the tumor remains unattainable, with residual cells driving recurrence that is resistant to conventional therapies. The GBM tumor microenviroment (TME) significantly impacts tumor progression and treatment response. In this review, we explore the emerging role of purinergic signaling, especially the P2X7 receptor (P2X7R). Due to its unique characteristics, it plays a key role in tumor progression and offers a potential therapeutic strategy for GBM through TME modulation. We discuss also the emerging role of the P2X4 receptor (P2X4R) as a promising therapeutic target. Overall, targeting purinergic signaling offers a potential approach to overcoming current GBM treatment limitations.
Collapse
Affiliation(s)
- Martina Bedeschi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.B.); (E.C.)
| | - Elena Cavassi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.B.); (E.C.)
| | - Antonino Romeo
- Radiation Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Anna Tesei
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.B.); (E.C.)
| |
Collapse
|
34
|
Jafari P, Forrest M, Segal J, Wang P, Tjota MY. Pan-Cancer Molecular Biomarkers: Practical Considerations for the Surgical Pathologist. Mod Pathol 2025; 38:100752. [PMID: 40058460 DOI: 10.1016/j.modpat.2025.100752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025]
Abstract
Traditional anatomic pathologic classification of cancer is based on tissue of origin and morphologic and immunohistochemical characterization of the malignant cells. With the technological improvements of massively parallel or next-generation sequencing, oncogenic drivers that are shared across different tumor types are increasingly being identified and used as pan-cancer biomarkers. This approach is reflected in the growing list of Food and Drug Administration-approved tumor-agnostic therapies, including pembrolizumab in the setting of microsatellite instability and high tumor mutational burden, larotrectinib and entrectinib for solid tumors with NTRK fusions, and combined dabrafenib-trametinib for BRAF V600E-mutated neoplasms. Several other biomarkers are currently under investigation, including fibroblast growth factor receptor (FGFR), RET, and ROS1 fusions; ERBB2 amplification; and mutations in the AKT1/2/3, NF1, RAS pathway and (mitogen-activated protein kinase (MAPK) pathway. As molecular assays are increasingly incorporated into routine tumor workup, the emergence of additional pan-cancer biomarkers is likely to be a matter more of "when" than "if." In this review, we first explore some of the conceptual and technical considerations at the intersection of surgical and molecular pathology, followed by a brief overview of both established and emerging molecular pan-cancer biomarkers and their diagnostic and clinical applications.
Collapse
Affiliation(s)
- Pari Jafari
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Megan Forrest
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Jeremy Segal
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Peng Wang
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | | |
Collapse
|
35
|
Alkayyal AA, Mahmoud AB. A 5-Year Update on the Clinical Development of Cancer Cell-Based Vaccines for Glioblastoma Multiforme. Pharmaceuticals (Basel) 2025; 18:376. [PMID: 40143152 PMCID: PMC11946125 DOI: 10.3390/ph18030376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Glioblastoma multiforme (GBM) is considered one of the most aggressive forms of brain cancer with a 15-month median survival, despite advancements in surgery, radiotherapy, and chemotherapy. The immune-suppressed tumor microenvironment and the blood-brain barrier are major contributors to its poor prognosis and treatment resistance. In the last decade, significant progress has been made in developing cell-based vaccines to boost immune responses against GBM. This review provides an extensive update on recent clinical trials involving various cancer cell vaccines, including ICT-107, the α-type-1 DC vaccine, and others. Although these trials have demonstrated potential improvements in progression-free survival (PFS) and overall survival (OS), the diverse and immune-suppressed nature of GBM poses challenges for consistent therapeutic success. We discuss the details of these trials along with the potential mechanism of vaccine efficacy and immune activations. The findings of these trials highlight the significance of a personalized immunotherapy approach and suggest that patient stratification could significantly advance the clinical management of GBM.
Collapse
Affiliation(s)
- Almohanad A. Alkayyal
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- College of Applied Medical Sciences, Taibah University, Madinah 41477, Saudi Arabia
- Health and Life Research Center, Taibah University, Madinah 42353, Saudi Arabia
| |
Collapse
|
36
|
Wiull K, Holmgren E, Svensson S, Eijsink VGH, Grönlund H, Mathiesen G. Surface display of two neoantigens on Lactiplantibacillus plantarum. Sci Rep 2025; 15:7769. [PMID: 40044906 PMCID: PMC11882890 DOI: 10.1038/s41598-025-91180-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: 09/02/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
Lactic acid bacteria, such as Lactiplantibacillus plantarum, are becoming increasingly popular hosts for combining production and delivery of therapeutic proteins to immune cells. Soluble antigens are susceptible to rapid proteolysis, hence anchoring of antigens to bacterial cells, which likely protects the antigen, is a preferred delivery strategy that may increase immune responses. In cancer research, personalized immunotherapy has high potential and, in this respect, the so-called neoantigens that accumulate in tumor cells are promising tumor specific targets. Here, we demonstrate that, when using the inducible pSIP expression system, L. plantarum can produce and surface display two neoantigens, NAG1 and ETV6. The antigens could be targeted to both the cell membrane and the cell wall, utilizing four different anchoring methods. The production level and the degree of surface exposure of the antigens varied among the anchors. Flow cytometry analysis showed that antigens anchored to the cell wall were more exposed than those anchored to the cell membrane. To our knowledge these are the first reports of neoantigens being produced and surface-displayed in Lactobacillales.
Collapse
Affiliation(s)
- Kamilla Wiull
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway.
| | - Erik Holmgren
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Vincent G H Eijsink
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway
| | - Hans Grönlund
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- NEOGAP Therapeutics AB, Stockholm, Sweden
| | - Geir Mathiesen
- Faculty of Chemistry, Biotechnology and Food Science, NMBU - Norwegian University of Life Sciences, Ås, Norway.
| |
Collapse
|
37
|
Luo S, Peng H, Shi Y, Cai J, Zhang S, Shao N, Li J. Integration of proteomics profiling data to facilitate discovery of cancer neoantigens: a survey. Brief Bioinform 2025; 26:bbaf087. [PMID: 40052441 PMCID: PMC11886573 DOI: 10.1093/bib/bbaf087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/29/2024] [Accepted: 02/19/2025] [Indexed: 03/10/2025] Open
Abstract
Cancer neoantigens are peptides that originate from alterations in the genome, transcriptome, or proteome. These peptides can elicit cancer-specific T-cell recognition, making them potential candidates for cancer vaccines. The rapid advancement of proteomics technology holds tremendous potential for identifying these neoantigens. Here, we provided an up-to-date survey about database-based search methods and de novo peptide sequencing approaches in proteomics, and we also compared these methods to recommend reliable analytical tools for neoantigen identification. Unlike previous surveys on mass spectrometry-based neoantigen discovery, this survey summarizes the key advancements in de novo peptide sequencing approaches that utilize artificial intelligence. From a comparative study on a dataset of the HepG2 cell line and nine mixed hepatocellular carcinoma proteomics samples, we demonstrated the potential of proteomics for the identification of cancer neoantigens and conducted comparisons of the existing methods to illustrate their limits. Understanding these limits, we suggested a novel workflow for neoantigen discovery as perspectives.
Collapse
Affiliation(s)
- Shifu Luo
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Hui Peng
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore
| | - Ying Shi
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- School of Computer and Information Technology, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Jiaxin Cai
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| | - Songming Zhang
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| | - Ningyi Shao
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Jinyan Li
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| |
Collapse
|
38
|
Song Z, Tao Y, You J. The potential applications of peptide-loading complex in cancer treatment. Front Immunol 2025; 16:1526137. [PMID: 40098955 PMCID: PMC11911339 DOI: 10.3389/fimmu.2025.1526137] [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: 11/11/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
Immunotherapy for cancer has made significant strides in the last several years. The prognosis for cancer patients has significantly improved as a result, particularly in hematological diseases. However, it was discovered that translating these achievements to solid tumors proved challenging. The peptide-loading complex (PLC), a temporary multisubunit membrane assembly in the endoplasmic reticulum (ER), is crucial for initiating a hierarchical immune response. Chaperones calreticulin and tapasin make up the PLC, unique to class I glycoproteins, thiooxido-reductase ERp57, and a transporter associated with antigen processing. The loading and editing of major histocompatibility complex class I (MHC-I) molecules with peptide translocation into the ER are synchronized by the PLC. One of the immune escape strategies revealed for tumors so far is changes in the expression of MHC molecules. This is because MHC antigens are crucial in presenting antigens to T-lymphocytes and controlling NK cell activity. Furthermore, decreased MHC-I expression has been linked to malignancies resistant to T-cell-based cancer immunotherapies (adoptive transfer of antitumor CD8 T-cells or checkpoint inhibition). The PLC is essential for T-cell priming, differentiation, and tumor growth control because it can bind to a wide range of MHC-I allomorphs. In this review, we have looked into PLC's function and effects in all forms of cancer to improve cancer therapy techniques.
Collapse
Affiliation(s)
- Zhidu Song
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Ying Tao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiaxin You
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
39
|
Bedi D, Hassan M, Yirsaw A, Vikas B, Datta P, Samuel T. The immunopeptidome of colon cancer cells treated with topoisomerase inhibiting drug reveals differential as well as common endogenous protein sampling and display of MHC I-associated peptides. Mol Cell Oncol 2025; 12:2471640. [PMID: 40051755 PMCID: PMC11881837 DOI: 10.1080/23723556.2025.2471640] [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: 01/16/2024] [Revised: 08/05/2024] [Accepted: 02/20/2025] [Indexed: 03/09/2025]
Abstract
Immunotherapy options for microsatellite stable (MSS) colorectal cancer are currently very limited. The lack of detectably unique or altered immunogens in the tumor microenvironment may be a factor. Radiation and chemotherapy may enhance immunotherapy by increasing cancer cell visibility through Major Histocompatibility Complex I (MHC I) expression. To investigate this, we treated MSS and microsatellite-instable (MSI) colon cancer cells with a topoisomerase inhibitor and analyzed MHC I-associated peptides. Treatment increased peptide numbers by 5% in RKO (MSI) cells and 83% in SW620 (MSS) cells, with 40-50% of peptides being exclusive to treatment. Additionally, clustering analysis revealed a set of peptides with uniquely conserved residues displayed only in treated MSS SW620 cells. Gene Ontology analysis of MHC I-displayed proteins revealed a treatment-induced increase in extracellular vesicle- and nuclear-derived proteins, alongside reduced cytosolic protein sampling. Overall, we present evidence for treatment-inducible differential display of peptides, some of which may affect interactions and functions of immune cells. Given the multitude of factors that modulate the effects of increased MHC I expression and associated peptides, further studies are needed to elucidate the pathophysiological implications of these changes.
Collapse
Affiliation(s)
- Deepa Bedi
- Departments of Pathobiology and Biomedical Sciences, Tuskegee University, College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL, USA
| | - Mohammed Hassan
- Departments of Pathobiology and Biomedical Sciences, Tuskegee University, College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL, USA
| | - Alehegne Yirsaw
- Departments of Pathobiology and Biomedical Sciences, Tuskegee University, College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL, USA
| | - Biba Vikas
- Departments of Pathobiology and Biomedical Sciences, Tuskegee University, College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL, USA
| | - Pran Datta
- University of Alabama at Birmingham, Birmingham, AL, USA
| | - Temesgen Samuel
- Departments of Pathobiology and Biomedical Sciences, Tuskegee University, College of Veterinary Medicine and Center for Biomedical Research, Tuskegee, AL, USA
| |
Collapse
|
40
|
Cao M, Yan J, Ding Y, Zhang Y, Sun Y, Jiang G, Zhang Y, Li B. The potential impact of RNA splicing abnormalities on immune regulation in endometrial cancer. Cell Death Dis 2025; 16:148. [PMID: 40032844 DOI: 10.1038/s41419-025-07458-7] [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: 10/08/2024] [Revised: 02/05/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025]
Abstract
RNA splicing controls the post-transcriptional level of gene expression, allowing for the synthesis of many transcripts with various configurations and roles. Variations in RNA splicing regulatory factors, including splicing factors, signaling pathways, epigenetic modifications, and environmental factors, are typically the origin of tumor-associated splicing anomalies. Furthermore, thorough literature assessments on the intricate connection between tumor-related splicing dysregulation and tumor immunity are currently lacking. Therefore, we also thoroughly discuss putative targets associated with RNA splicing in endometrial cancer (EC) and the possible impacts of aberrant RNA splicing on the immune control of tumor cells and tumor microenvironment (TME), which contributes to enhancing the utilization of immunotherapy in the management of EC and offers an alternative viewpoint for the exploration of cancer therapies and plausible prognostic indicators.
Collapse
Affiliation(s)
- Minyue Cao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jiayu Yan
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yan Ding
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yiqin Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yihan Sun
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Genyi Jiang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yanli Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Bilan Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
41
|
Luo M, Yang J, Schäffer AA, Chen C, Liu Y, Chen Y, Lin C, Diao L, Zang Y, Lou Y, Salman H, Mills GB, Ruppin E, Han L. Ancestral Differences in Anticancer Treatment Efficacy and Their Underlying Genomic and Molecular Alterations. Cancer Discov 2025; 15:511-529. [PMID: 39601595 PMCID: PMC11875934 DOI: 10.1158/2159-8290.cd-24-0827] [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: 06/10/2024] [Revised: 09/12/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024]
Abstract
SIGNIFICANCE Our study charts a global landscape of ancestry-associated differences in therapeutic efficacy, highlighting the importance of considering ancestry in anticancer therapies.
Collapse
Affiliation(s)
- Mei Luo
- Brown Center for Immunotherapy, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jingwen Yang
- Brown Center for Immunotherapy, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alejandro A. Schäffer
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Chengxuan Chen
- Brown Center for Immunotherapy, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yuan Liu
- Brown Center for Immunotherapy, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yamei Chen
- Brown Center for Immunotherapy, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chunru Lin
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lixia Diao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yong Zang
- Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Huda Salman
- Brown Center for Immunotherapy, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Division of Hematology-Oncology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gordon B. Mills
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Leng Han
- Brown Center for Immunotherapy, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Department of Biostatistics and Health Data Science, School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
42
|
Mi Y, Jiang P, Luan J, Feng L, Zhang D, Gao X. Peptide‑based therapeutic strategies for glioma: Current state and prospects. Peptides 2025; 185:171354. [PMID: 39922284 DOI: 10.1016/j.peptides.2025.171354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Glioma is a prevalent form of primary malignant central nervous system tumor, characterized by its cellular invasiveness, rapid growth, and the presence of the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB). Current therapeutic approaches, such as chemotherapy and radiotherapy, have shown limited efficacy in achieving significant antitumor effects. Therefore, there is an urgent demand for new treatments. Therapeutic peptides represent an innovative class of pharmaceutical agents with lower immunogenicity and toxicity. They are easily modifiable via chemical means and possess deep tissue penetration capabilities which reduce side effects and drug resistance. These unique pharmacokinetic characteristics make peptides a rapidly growing class of new therapeutics that have demonstrated significant progress in glioma treatment. This review outlines the efforts and accomplishments in peptide-based therapeutic strategies for glioma. These therapeutic peptides can be classified into four types based on their anti-tumor function: tumor-homing peptides, inhibitor/antagonist peptides targeting cell surface receptors, interference peptides, and peptide vaccines. Furthermore, we briefly summarize the results from clinical trials of therapeutic peptides in glioma, which shows that peptide-based therapeutic strategies exhibit great potential as multifunctional players in glioma therapy.
Collapse
Affiliation(s)
- Yajing Mi
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Pengtao Jiang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Jing Luan
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lin Feng
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Dian Zhang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Xingchun Gao
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China.
| |
Collapse
|
43
|
Khaddour K, Buchbinder EI. Individualized Neoantigen-Directed Melanoma Therapy. Am J Clin Dermatol 2025; 26:225-235. [PMID: 39875711 DOI: 10.1007/s40257-025-00920-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2025] [Indexed: 01/30/2025]
Abstract
Individualized neoantigen-directed therapy represents a groundbreaking approach in melanoma treatment that leverages the patient's own immune system to target cancer cells. This innovative strategy involves the identification of unique immunogenic neoantigens (mutated proteins specific to an individual's tumor) and the development of therapeutic vaccines that either consist of peptide sequences or RNA encoding these neoantigens. The goal of these therapies is to induce neoantigen-specific immune responses, enabling the immune system to recognize and destroy cancer cells presenting the targeted neoantigens. This individualized approach is particularly advantageous given the genetic heterogeneity of melanoma, which exhibits distinct mutations among different patients. In contrast to traditional therapies, neoantigen-directed therapy offers a tailored treatment that potentially reduces off-target side effects and enhances therapeutic efficacy. Recent advances in neoantigen prediction and vaccine development have facilitated clinical trials exploring the combination of neoantigen vaccines with immune checkpoint inhibitors. These trials have shown promising clinical outcomes, underscoring the potential of this personalized approach. This review provides an overview of the rationale behind neoantigen-directed therapies and summarizes the current state of knowledge regarding personalized neoantigen vaccines in melanoma treatment.
Collapse
Affiliation(s)
- Karam Khaddour
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA.
- Melanoma Disease Center, Dana-Farber Cancer Institute, Boston, MA, 02115, USA.
- Harvard Medical School, Boston, MA, 02115, USA.
| | - Elizabeth I Buchbinder
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Melanoma Disease Center, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
44
|
Rojas-Domínguez A, Martínez-Vargas IU, Alvarado-Mentado M. Modeling and simulation of genotypic Tumor Mutational Burden and Phenotypic Immunogenicity biomarkers in cancer immunoediting with Ising-Hamiltonian characterization. Comput Biol Med 2025; 187:109717. [PMID: 39894008 DOI: 10.1016/j.compbiomed.2025.109717] [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: 08/08/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND AND OBJECTIVE In the Tumor Micro-Environment, cancer progression and its relationship with the Immune System (IS) are described in terms of cancer immunoediting (CI) phases, each of which is characterized by different types and levels of interaction between the tumor cells and elements of the IS, such as CD8+T cells. Said interactions are governed by genotypical (Tumor Mutational Burden, TMB) and phenotypical aspects pertaining to the tumor, as well as by the strength of the IS. In this work, a computational model of CI is presented that incorporates the TMB and the biomarker Tumor Immunogenic Phenotype (TIP) as its control parameters, and which employs the Ising-model Hamiltonian to characterize the system with respect to the CI phases. METHODS Our model is a probabilistic multi-agent system with agents for tumor cells and for the IS. The computer implementation includes the parametrization of the TMB and the TIP, which is useful for identifying whether a tumor is hot or cold based on tumor immunogenicity and inflammation. For modeling the interactions between tumor and immune cells, the relevant elements are integrated under a Michaelis-Menten equation that regulates the recruitment rate of CD8+T cells and other IS elements. This novel quantification of immune cell recruitment encompasses the growth of neoantigen production, which in turn triggers the growth of CD8+T cells. RESULTS Our model reliably captures the Elimination, Equilibrium, and Escape phases of tumor-immune cell interactions, modulating the observed behaviors through the introduced parametrization of TMB and TIP biomarkers. Notably, these results align well with the combination of genotypical and phenotypical biomarkers analyzed in recent literature. A remarkable instance is the appreciable inhibition of the tumor activity during the Escape phase, observed for phenotypically hot tumors with relatively high TMB, and pointing towards improved efficacy of the IS against such tumors. The Ising-Hamiltonian provides precise quantification of diverse tumor-immune interactions across different TMB and TIP value combinations. CONCLUSIONS The presented model, formed by relatively simple agents, generates emergent behaviors through which the phases of CI are identified. The flexible choice of control parameters is robust enough and provides a plausible explanation for the mechanisms through which tumors with high TMB and high immunogenicity (i.e., hot tumors) exhibit a higher probability of responding to immunotherapy treatment. Characterization via the Ising-model Hamiltonian supports this explanation by summarizing the system's dynamics, which, in turn, facilitates its analysis and methodical improvements. The complex interplay of TMB, TIP, and individual physiology is finely captured.
Collapse
Affiliation(s)
- Alfonso Rojas-Domínguez
- Centro de Investigación en Computación-IPN, Av. Juan de Dios Bátiz, esq. Miguel Othón de Mendizábal, Col.Nueva Industrial Vallejo, CDMX, 0700, Mexico
| | - Irving Ulises Martínez-Vargas
- Depto. de Computación CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco., CDMX, 07360, Mexico; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place., New York, 10029, NY, USA
| | - Matías Alvarado-Mentado
- Depto. de Computación CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco., CDMX, 07360, Mexico.
| |
Collapse
|
45
|
Martel-Martel A, Sinha KM, Vilar E. Neoantigen Vaccines in Cancer Prevention. Cancer J 2025; 31:e0763. [PMID: 40126885 DOI: 10.1097/ppo.0000000000000763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 03/26/2025]
Abstract
Recent advances in cancer immunotherapy have established neoantigen-based vaccines as a promising approach to cancer prevention. Unlike tumor-associated antigens, neoantigens originate exclusively from somatic mutations, thus enabling tumor-specific targeting without harm to normal tissues. This distinctive feature promotes robust immune responses while reducing the risk of autoimmune side effects. Developing standardized "off-the-shelf" vaccines targeting shared neoantigens offers a scalable strategy for cancer prevention, particularly benefitting genetically predisposed high-risk populations. These vaccines can be administered to high-risk individuals before malignant transformation to potentially intercept cancer development through early immune activation. Advances in next-generation sequencing and computational biology have increased the accuracy of neoantigen prediction, while advances in vaccine delivery platforms have boosted vaccine efficacy. The integration of neoantigen-based vaccines with immune checkpoint inhibitors, immune stimulants, and classic chemopreventive agents has a synergistic potential to improve cellular immunity. This review examines biological mechanisms, clinical development, and future directions of neoantigen-based vaccines in cancer prevention, emphasizing their clinical potential to revolutionize risk-reduction strategies.
Collapse
Affiliation(s)
- Abel Martel-Martel
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | |
Collapse
|
46
|
Ninmer EK, Xu F, Slingluff CL. The Landmark Series: Cancer Vaccines for Solid Tumors. Ann Surg Oncol 2025; 32:1443-1452. [PMID: 39704984 PMCID: PMC11811251 DOI: 10.1245/s10434-024-16712-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/01/2024] [Indexed: 12/21/2024]
Abstract
Immunotherapy has become an integral part of the treatment for solid tumors. Cancer vaccines represent a potentially powerful class of immunotherapeutic agents to drive antitumor immunity. Cancer vaccine development involves selecting immunogenic target antigens expressed by tumor cells that can be effectively delivered for uptake by antigen-presenting cells to generate a robust adaptive immune response against tumor. While numerous cancer vaccines have been shown to produce antigen-specific immune responses, translating promising results of immunogenicity from early-phase trials into durable clinical benefit in larger randomized trials has remained elusive. Recent findings support new enthusiasm for several cancer vaccine approaches for solid tumors. This review will discuss landmark historic clinical trials in cancer vaccine development and strategies to optimize cancer vaccines to achieve improved clinical efficacy.
Collapse
Affiliation(s)
- Emily K Ninmer
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Feifan Xu
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Craig L Slingluff
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA.
- School of Medicine, Cancer Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
47
|
Klein J, Tran WT, Viswanathan S, Salgado R, Poortmans P, Machiels M. Tumour-infiltrating Lymphocytes and Radiation Therapy in Rectal Cancer: Systematic Review and Meta-analysis. Clin Oncol (R Coll Radiol) 2025; 39:103742. [PMID: 39854781 DOI: 10.1016/j.clon.2024.103742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025]
Abstract
AIM Tumour-infiltrating lymphocytes (TILs) represent a promising cancer biomarker. Different TILs, including CD8+, CD4+, CD3+, and FOXP3+, have been associated with clinical outcomes. However, data are lacking regarding the value of TILs for patients receiving radiation therapy (RT). We conducted a systemic review and meta-analysis of available data evaluating TILs for patients receiving curative-intent therapy including RT. MATERIALS AND METHODS Eligible studies presented a defined cohort of patients who all received curative-intent therapy, including RT, and also reported the relationship between any TIL score and either tumour response or survival outcomes. After comprehensive search of online databases (PubMed, EMBASE, Cochrane, and Web of Science), 2 authors conducted title, abstract, and whole-text review for quality and risk of bias following Preferred Reporting Items for Systematic Review and Meta-Analyses (PRISMA) guidelines. Data from publications that met quality criteria were grouped via (1) TIL analysed, (2) pre- or post-RT TIL assessment, and (3) clinical outcome measured. RESULTS Initial search yielded 669 unique studies. Thirty-one studies met quality criteria, of which 20 studied rectal cancer (RC), 4 oesophageal, 3 pancreas, 2 lung, cervical/uterine 1 each. We conducted systematic review and meta-analysis of the RC publications. All except 2 were single-institutional cohort studies. After meta-analysis, the pre-RT epithelial CD8+ (p = 0.04) and stromal FOXP3+ (p = 0.01) counts were associated with survival without disease, while pre-RT epithelial (p = 0.02) and stromal (p = 0.001) FOXP3+ TILs were associated with overall survival. On post-RT analysis, epithelial (p = .04) and stromal (p = 0.02) CD8+ TILs were associated with survival without disease and epithelial CD8+ TILs were associated with overall survival (p = 0.01).Preoperative CD8+ and FOXP3+ TILs were generally associated with tumour response to RT, but meta-analysis was not conducted due to heterogeneity of response measurement techniques. CONCLUSION TILs represent a useful parameter for tumour response and survival outcomes for patients receiving curative-intent therapy, including RT for RC. Future work should aim to standardise TIL measurement and quantification methods and to develop protocols to clarify clinical application of these findings.
Collapse
Affiliation(s)
- J Klein
- Department of Radiation Oncology, State University of New York (SUNY) Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY, USA; Department of Radiation Oncology, Maimonides Medical Center, 6300 8th Ave, Brooklyn, NY, USA.
| | - W T Tran
- Department of Radiation Oncology, University of Toronto, 149 College St#504, Toronto, Ontario, Canada; Sunnybrook Research Institute, 2075 Bayview Ave, Toronto, Ontario, Canada.
| | - S Viswanathan
- Department of Epidemiology and Population Health, Montefiore Einstein Comprehensive Cancer Center, 1300 Morris Park Ave, Block Building Room 315, Bronx, NY, USA.
| | - R Salgado
- Division of Research, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Australia; Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium.
| | - P Poortmans
- Faculty of Medicine and Health Sciences, University of Antwerp, Prinsstraat 13, 2000, Antwerp, Belgium; Department of Radiation Oncology, Iridium Netwerk, Oosterveldlaan 22, 2610, Antwerp, Belgium.
| | - M Machiels
- Faculty of Medicine and Health Sciences, University of Antwerp, Prinsstraat 13, 2000, Antwerp, Belgium; Department of Radiation Oncology, Iridium Netwerk, Oosterveldlaan 22, 2610, Antwerp, Belgium.
| |
Collapse
|
48
|
Li D, Andaloori L, Crowe M, Lin S, Hong J, Zaidi N, Ho M. Development of CAR-T Therapies and Personalized Vaccines for the Treatment of Cholangiocarcinoma: Current Progress, Mechanisms of Action, and Challenges. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:453-469. [PMID: 39675505 DOI: 10.1016/j.ajpath.2024.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 10/15/2024] [Accepted: 10/24/2024] [Indexed: 12/17/2024]
Abstract
Cholangiocarcinoma (CCA) is a highly fatal malignancy with an increasing prevalence, a high mortality rate, poor overall survival, and limited responsiveness to conventional chemoradiotherapy. Targeted therapies addressing specific gene mutations have expanded treatment options for some patient populations. The introduction of chimeric antigen receptor-modified T-cell (CAR-T) immunotherapy and personalized vaccines have opened up a new avenue for managing various cancers. Considerable efforts have been dedicated to preclinical research and ongoing clinical trials of immunotherapeutic approaches including CAR-T therapy, vaccines, and antibody-based therapies such as antibody drug conjugates. However, the potential of CAR-T therapy and vaccines in treating advanced unresectable/metastatic cholangiocarcinoma remains largely unexplored. This article offers an overview of the current landscape of antibody-based immunotherapy, particularly CAR-T therapy and vaccines in the context of cholangiocarcinoma treatment. It outlines a framework for selecting CAR-T and vaccine targets and delves into the biology of promising targetable antigens, as well as potential future therapeutic targets.
Collapse
Affiliation(s)
- Dan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Lalitya Andaloori
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Matthew Crowe
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Shaoli Lin
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jessica Hong
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Neeha Zaidi
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland.
| | - Mitchell Ho
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
49
|
Pandey K, Faridi P, Ayala R, Lee YCG, Rouse E, Krishna SSG, Dick I, Redwood A, Robinson B, Creaney J, Purcell AW. Multiple Classes of Antigen Contribute to the Antigenic Landscape of Mesothelioma. Mol Cell Proteomics 2025; 24:100925. [PMID: 39921204 PMCID: PMC11929013 DOI: 10.1016/j.mcpro.2025.100925] [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: 08/04/2024] [Revised: 01/23/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025] Open
Abstract
Mesothelioma is an incurable, asbestos-exposure-related cancer that typically affects the lining or pleura of the lungs. Symptoms typically develop many decades after initial asbestos exposure, leaving an enduring legacy of disease. The current disease burden is peaking worldwide and thus there is a massive unmet clinical need for curative therapies. Recently, immune checkpoint blockade-based therapy has been adopted as a first-line of treatment for mesothelioma. Vaccine-induced augmentation of immune responses unleashed during checkpoint blockade may provide further clinical benefit in mesothelioma. In this study, we explore the human leukocyte antigen class I landscape (or immunopeptidome) of mesothelioma in patient-derived cell lines and clinical material (pleural effusion samples). We identify a range of peptide antigens derived from targets including cancer testis antigens, endogenous retroviruses as well as novel post-translational modification of peptides. This information will facilitate the characterization of the immune response to these antigens to determine which class of antigen is most immunogenic and has the potential to be tested in future vaccine studies.
Collapse
Affiliation(s)
- Kirti Pandey
- Department of Biochemistry and Molecular Biology and Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Pouya Faridi
- Department of Medicine, School of Clinical Sciences, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia
| | - Rochelle Ayala
- Department of Biochemistry and Molecular Biology and Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Y C Gary Lee
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Australia; Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Australia
| | - Ebony Rouse
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Australia
| | - Sanjay S G Krishna
- Department of Biochemistry and Molecular Biology and Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Ian Dick
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Australia; Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Australia
| | - Alec Redwood
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Australia; Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Australia
| | - Bruce Robinson
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Australia; Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Australia
| | - Jenette Creaney
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Australia; Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Australia.
| | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology and Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.
| |
Collapse
|
50
|
Baharom F, Hermans D, Delamarre L, Seder RA. Vax-Innate: improving therapeutic cancer vaccines by modulating T cells and the tumour microenvironment. Nat Rev Immunol 2025; 25:195-211. [PMID: 39433884 DOI: 10.1038/s41577-024-01091-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 10/23/2024]
Abstract
T cells have a critical role in mediating antitumour immunity. The success of immune checkpoint inhibitors (ICIs) for cancer treatment highlights how enhancing endogenous T cell responses can mediate tumour regression. However, mortality remains high for many cancers, especially in the metastatic setting. Based on advances in the genetic characterization of tumours and identification of tumour-specific antigens, individualized therapeutic cancer vaccines targeting mutated tumour antigens (neoantigens) are being developed to generate tumour-specific T cells for improved therapeutic responses. Early clinical trials using individualized neoantigen vaccines for patients with advanced disease had limited clinical efficacy despite demonstrated induction of T cell responses. Therefore, enhancing T cell activity by improving the magnitude, quality and breadth of T cell responses following vaccination is one current goal for improving outcome against metastatic tumours. Another major consideration is how T cells can be further optimized to function within the tumour microenvironment (TME). In this Perspective, we focus on neoantigen vaccines and propose a new approach, termed Vax-Innate, in which vaccination through intravenous delivery or in combination with tumour-targeting immune modulators may improve antitumour efficacy by simultaneously increasing the magnitude, quality and breadth of T cells while transforming the TME into a largely immunostimulatory environment for T cells.
Collapse
Affiliation(s)
| | - Dalton Hermans
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
| | | | - Robert A Seder
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|