免疫检查点阻断(ICB)疗法
Search documents
深圳湾实验室×北京大学合作最新Nature:陈鹏/席建忠合作开发全新癌症疫苗——瘤内疫苗嵌合体
生物世界· 2026-01-08 04:33
撰文丨王聪 编辑丨王多鱼 排版丨水成文 肿瘤通过多种机制降低免疫原性并逃逸免疫监视,包括过表达抑制性检查点蛋白、新抗原编辑以及抗原呈 递功能受损。恢复抗肿瘤免疫从根本上依赖于通过增强肿瘤抗原呈递来补充和激活肿瘤反应性 T 细胞。 在 陈鹏 团队前期系统性构建 膜蛋白靶向降解 (m eTPD ) 技术体系的基础上,研究团队创新性地提出了 将 蛋白降解 与 抗原呈递通路 深度耦合 的策略,利用 meTPD 技术,开发出了同时实现 " 免疫检查点降解 " 和 "高质量 抗原呈递 "的双功能 " 瘤内疫苗嵌合体 " ( Intratumoural vaccination chimera, iVAC ) , 这种以肿瘤为出发点的" 降解型疫苗 "为攻克癌症的 免疫 耐受提供了新的范式。 免疫检查点阻断 (ICB) 疗法革新了癌症治疗,但由于肿瘤特异性细胞毒性 T 淋巴细胞 (CTL) 的缺乏 或功能障碍,其应答率仍然有限。虽然过继 T 细胞转移和癌症疫苗已被设计用于增加 CTL 数量并增强抗肿 瘤反应,但疫苗诱导的 CTL 常常在肿瘤微环境 (TME) 的免疫抑制条件下失效,因此,需要与其他疗法 联合使用。 一种能在生 ...
南方医科大学最新Cell子刊:揭示尿苷调控CD8⁺ T细胞抗肿瘤免疫新机制
生物世界· 2025-12-31 04:34
免疫检查点阻断 ( ICB ) 疗法,虽在癌症治疗中取得显著临床效益,但面临治疗耐受性和适应症不明确 等局限,影响了其广泛应用。 肿瘤微环境中的代谢产物对 ICB 治疗抵抗发挥重要作用, 尿苷 作为肿瘤细 胞代谢产物,具有维持细胞能量、促进 ATP 合成及参与糖蛋白合成 (例如 UDP-GlcNAc ) 等多重功能 。研究表明,尿苷可通过促进淋巴细胞分化增强抗感染免疫力 。然而,尿苷调控 T 细胞抗肿瘤免疫的作用 和分子机制,目前仍不清楚。 2025 年 12 月 29 日,南方医科大学基础医学院病理学系 / 南方医院病理科 梁莉 、 王志章 教授团队 ( 肖建彪、李志洋、丁轶、朱科锦、郑志浩为论文共同第一作者 ) , 在 Cell Metabolism 期刊 发表了题 为: Uridine Depletion Impairs CD8⁺ T Cell Antitumor Activity through N-Glycosylation 的研究论文。 编辑丨王多鱼 排版丨水成文 该研究揭示了 尿苷 介导 CD45 蛋白的 N- 糖基化促进 CD8⁺ T 细胞抗肿瘤免疫 的新机制,系统阐述了肿 瘤微环境内尿苷耗竭 ...
Science子刊:卞修武团队发现增强CAR-T细胞抗肿瘤效果的基因修饰策略
生物世界· 2025-10-25 08:00
Core Viewpoint - CAR-T cell therapy has shown significant efficacy in treating hematological malignancies, prompting research into its application for solid tumors, particularly glioblastoma multiforme (GBM), which presents unique treatment challenges due to its aggressive nature and lack of effective therapies [2][4]. Group 1: T Cell Exhaustion and Mechanisms - T cell exhaustion is a major barrier to the efficacy of CAR-T cell therapy in solid tumors, characterized by reduced proliferation, impaired effector function, and increased expression of inhibitory receptors [2][4]. - Recent advancements in single-cell RNA sequencing (scRNA-seq) have provided insights into the molecular mechanisms of T cell exhaustion, identifying key regulatory factors such as DNMT3A, SOX4, and PRDM1 that limit T cell anti-tumor activity [2][4]. Group 2: Research Findings on NR4A3 and FOS - A study published in Science Advances found that knocking down NR4A3 enhances CAR-T cell efficacy against malignant gliomas, but this effect diminishes due to T cell exhaustion induced by chronic antigen exposure [3][5]. - Enhancing FOS expression in NR4A3-deficient CAR-T cells can reverse T cell functional exhaustion, thereby maintaining tumor clearance capabilities and improving therapeutic efficacy [3][5][6]. Group 3: Implications for CAR-T Cell Therapy - The research highlights the critical role of NR4A3 in regulating T cell cytotoxicity and memory formation during early antigen exposure, suggesting a combined genetic modification strategy of NR4A3 knockdown and FOS overexpression to protect CAR-T cells from exhaustion [6][8]. - This dual modification approach could lead to sustained tumor clearance in solid tumors, offering a promising new strategy for optimizing CAR-T cell therapy in clinical settings [5][6].
四川大学最新Cell子刊论文:仿生纳米生物催化剂,让冷肿瘤变热,增强免疫治疗效果
生物世界· 2025-09-14 04:05
Core Viewpoint - Immunotherapy, particularly immune checkpoint blockade (ICB), has transformed cancer treatment but remains ineffective in "cold tumors" due to immune suppression in the tumor microenvironment (TME) [2][5][6] Group 1: Research Findings - A new biomimetic Ru/TiO₂ nanobiocatalyst system inspired by natural enzyme reaction systems (ERS) has been developed, capable of rapid, pH-dependent generation of reactive oxygen species (ROS) and oxygen (O₂), effectively converting cold tumors into hot tumors [3][6][7] - The Ru/TiO₂ system enhances anti-tumor immunity and suppresses tumor metastasis when used in conjunction with ICB therapy [3][7] - This research establishes a precedent for adaptive nanobiocatalysts in the TME and paves the way for the development of next-generation immunotherapies targeting drug-resistant cancers [3][6] Group 2: Mechanism of Action - The study demonstrates that Ru/TiO₂ can mediate immunogenic cell death (ICD) in melanoma cells through endoplasmic reticulum stress, while also inhibiting hypoxia-induced immune suppression [7] - The design of Ru/TiO₂ aims to reverse immune suppression and enhance immunogenicity, transforming "immune cold" tumors into "immune hot" tumors [7] Group 3: Clinical Implications - The findings suggest that the rational design of robust and efficient biocatalytic materials could extend beyond cancer treatment, opening new avenues for immune modulation in other diseases [3][6]
北京大学最新Cell子刊:激活肿瘤细胞焦亡,提高抗肿瘤免疫效果
生物世界· 2025-09-06 04:05
Core Viewpoint - Tumor immunotherapy, particularly immune checkpoint blockade (ICB) targeting the PD-1/PD-L1 pathway, shows significant promise in treating various advanced cancers, but low immune response rates hinder its efficacy and widespread application [2] Group 1: Research Findings - The study developed a self-luminous nanosystem that enhances the activation of pyroptosis in tumor cells, leading to a strong antitumor immune response when combined with anti-PD-L1 monoclonal antibodies [3][6] - Pyroptosis, a newly discovered form of immunogenic cell death (ICD), releases pro-inflammatory cytokines and damage-associated molecular patterns, triggering a robust antigen-specific immune response [5] - The self-luminous nanoparticles can emit light within the tumor without the need for an external light source, enhancing the generation of reactive oxygen species (ROS) and achieving significant tumor-killing effects [7] Group 2: Mechanism and Components - The nanosystem consists of amphiphilic porphyrin lipids, camptothecin derivatives, and a targeting moiety, which together facilitate the release of oxygen and hydrogen peroxide in the acidic tumor microenvironment [6] - The combination of chemotherapy and self-enhanced photodynamic therapy synergistically activates pyroptosis, driving immune activation that enhances the antitumor response to PD-L1 therapy [7]
西南交通大学发表最新Cell子刊论文
生物世界· 2025-08-25 10:30
Core Viewpoint - The article discusses the development of a hydrogel-fiber composite device (HFCD) aimed at enhancing the efficacy of immune checkpoint blockade (ICB) therapy for recurrent glioblastoma (GBM) by activating cytotoxic T lymphocytes (CTL) in the postoperative tumor microenvironment (pTME) [2][3][5]. Group 1: Background on Glioblastoma and ICB Therapy - Glioblastoma (GBM) is the most common and aggressive brain tumor, accounting for approximately 57% of all gliomas, with a recurrence rate exceeding 90% due to its invasive nature [5]. - The postoperative tumor microenvironment (pTME) is characterized by immune suppression, including CTL exhaustion and infiltration of immunosuppressive cells, which reduces the clinical benefits of ICB therapy [5][6]. - There is a need to reshape the immune landscape within the pTME to enhance the response and durability of ICB therapy against GBM recurrence [5][6]. Group 2: Development of HFCD - The HFCD is designed to locally activate CTLs and modulate the acidic pTME, creating a favorable niche for CTLs [3][8]. - The device releases chemokine CXCL10 and PD-L1 inhibitors in a timed manner, enhancing CTL infiltration and maintaining their cytotoxic function [3][8]. - In an in situ GBM resection model, HFCD treatment achieved a 40% rate of complete recurrence inhibition and significantly extended the median survival to 49 days [9][10]. Group 3: Mechanism of Action - HFCD consists of quaternized chitosan hydrogel and electrospun fibers, which neutralize the acidic pTME and release CXCL10 to recruit CTLs [8]. - The sustained release of PD-L1 inhibitors from the PLGA matrix maintains PD-L1 blockade, enhancing CTL recognition and cytotoxic activity against residual GBM cells [8][10]. - This strategy alleviates immune suppression in the GBM pTME and enhances the protective effect of ICB against GBM recurrence [8][10].
Cell子刊:浙江大学周民团队开发药食同源策略,增强抗肿瘤免疫的同时降低毒副作用
生物世界· 2025-08-25 08:10
Core Viewpoint - The research highlights the importance of gut microbiota in enhancing the efficacy and safety of immune checkpoint blockade (ICB) therapy for cancer treatment, proposing a novel food-medicine homologous formula to improve outcomes and reduce adverse effects [2][6][10]. Group 1: Research Development - A new oral formulation, CV/APS-MS, was developed using microcapsules to co-load Chlorella vulgaris and Astragalus polysaccharides, which are recognized for their therapeutic and nutritional benefits [3][6]. - This formulation aims to prolong retention time in the gut, nourish beneficial gut microbiota, and alleviate inflammation [6][8]. Group 2: Experimental Findings - In mouse models of melanoma lung metastasis treated with ICB therapy, CV/APS-MS improved T cell-mediated anti-tumor immunity and mitigated ICB-induced colitis and pneumonia by restoring gut microbiota balance and reducing pro-inflammatory cytokines [8][10]. - The study suggests that combining food-grade bioreagents with modern medicine could be a powerful method to enhance cancer treatment efficacy and tolerance [10].
登上Cell子刊封面,中国药科大学揭示酪氨酸激酶抑制剂通过肠道菌群增强癌症免疫疗法
生物世界· 2025-06-22 03:38
Core Viewpoint - The study highlights the role of gut microbiota, specifically the metabolite urocanic acid (UCA), in enhancing the efficacy of cancer immunotherapy when combined with tyrosine kinase inhibitors (TKIs) [3][8][11]. Group 1: Research Findings - The research demonstrates that TKIs increase the abundance of the gut bacterium Muribaculum gordoncarteri and its metabolite UCA, which enhances the response to immune checkpoint blockade (ICB) therapy [8][9]. - UCA interacts with IκBα to inhibit NF-κB activation in endothelial cells, thereby reducing the recruitment of myeloid-derived suppressor cells (MDSCs) mediated by CXCL1 [9][11]. - Higher levels of UCA and Muribaculum gordoncarteri are found in the feces of patients who respond to ICB therapy compared to non-responders, suggesting their potential as predictive biomarkers for treatment response [8][9][11]. Group 2: Implications for Cancer Treatment - The findings indicate that the interaction between TKIs and gut microbiota could be a crucial factor in improving cancer treatment outcomes, particularly for patients who currently do not respond well to existing therapies [7][9]. - Understanding the mechanisms by which UCA enhances ICB therapy could lead to new strategies for increasing the effectiveness of cancer immunotherapy [3][11].
Nature:华人团队开发新型PROTAC,治疗多种癌症类型,一作将回国加入南京大学
生物世界· 2025-05-27 03:57
Core Viewpoint - Immune checkpoint blockade (ICB) therapies, represented by anti-PD-1 and anti-PD-L1 monoclonal antibodies, have significantly transformed cancer treatment, yet many patients show poor response or develop resistance to these therapies [2][6]. Group 1: Research Findings - A study published in Nature by a team from the University of Michigan reveals that the balance between STAT5 and STAT3 shapes dendritic cell (DC) function and tumor immunity, leading to the development of a STAT3-targeting PROTAC that enhances tumor sensitivity to ICB therapy [3][10]. - The research indicates that the limited number and impaired function of dendritic cells in the tumor microenvironment (TME) hinder the effectiveness of ICB therapies, emphasizing the need to understand the mechanisms behind dendritic cell phenotype formation [6][8]. Group 2: Mechanisms of Action - STAT3 is often activated in the TME, mediating immune suppression and promoting tumor growth factors, while STAT5 is activated by cytokine signals and plays a positive role in anti-tumor immune responses [7][9]. - The study found that ICB therapy reprograms the interaction between STAT3 and STAT5 pathways in dendritic cells, activating T cell immunity and enhancing the efficacy of ICB [9][10]. Group 3: Therapeutic Implications - The development of STAT3 degradation agents, such as SD-36 and SD-2301, shows promise in reprogramming dendritic cells towards an immunogenic state, effectively treating advanced tumors and those resistant to ICB therapy without toxicity [9][10]. - This research opens new avenues for cancer immunotherapy by targeting the dynamic balance between STAT3 and STAT5 in dendritic cells [10].