Workflow
癌症免疫治疗
icon
Search documents
Nature子刊:IL-10-mRNA 纳米颗粒,重编程肿瘤免疫,提高癌症免疫治疗效果并防止复发
生物世界· 2025-08-19 04:04
Core Viewpoint - The research led by Professor Tao Wei from Harvard Medical School demonstrates that IL-10-mRNA nanoparticles can significantly enhance cancer immunotherapy effectiveness and induce lasting anti-tumor immune memory, effectively preventing cancer recurrence [2][3][5]. Group 1: Research Findings - The study confirms that intravenous injection of IL-10-mRNA nanoparticles triggers strong immune surveillance in various preclinical tumor models while reducing systemic toxicity [4]. - IL-10-mRNA nanoparticles maintain local IL-10 production, promoting extensive infiltration and proliferation of cytotoxic T cells, activation and maturation of dendritic cells, and upregulation of MHC-I molecules in immunosuppressive early-stage liver cancer [5]. - In a model of advanced liver cancer, the combination of IL-10-mRNA nanoparticles and immune checkpoint blockade therapy achieved complete tumor clearance in 43% of mice, extending median survival sixfold compared to the immune checkpoint blockade alone, and providing 100% protection against tumor reattack [5][7]. Group 2: Implications for Treatment - The strategy of intravenous injection of IL-10-mRNA nanoparticles is expected to overcome the limitations faced by recombinant IL-10 in clinical trials for various immunosuppressive tumors [7].
Cancer Cell:朱波/刘新东/贾罄竹团队发现癌症治疗新靶点——DNASE1L3
生物世界· 2025-08-15 04:21
Core Viewpoint - The study highlights the role of DNASE1L3-expressing dendritic cells in enhancing CD8+ T cell function and improving the efficacy of anti-PD-1/PD-L1 therapies by degrading neutrophil extracellular traps (NETs) [2][3][5]. Group 1: Research Findings - The expression of DNASE1L3 in tumor-infiltrating dendritic cells is positively correlated with better prognosis in cancer patients undergoing anti-PD-1/PD-L1 therapy [5]. - Conditional knockout of DNASE1L3 in dendritic cells leads to accelerated tumor growth and reduced efficacy of anti-PD-L1 therapy due to impaired CD8+ T cell infiltration and function [5]. - Exogenous supplementation of DNASE1L3 enhances CD8+ T cell infiltration into the tumor microenvironment, reduces T cell exhaustion, significantly inhibits tumor growth, and improves responses to anti-PD-L1 therapy [5]. Group 2: Mechanistic Insights - DNASE1L3+ dendritic cells maintain a cytotoxic CD8+ T cell hub by degrading NETs, which inhibit the spatial distribution of CD8+ T cells within tumors [5][8]. - The absence of DNASE1L3 in dendritic cells promotes tumor growth through CD8+ T cell dysfunction [5][8]. Group 3: Implications for Therapy - DNASE1L3 is identified as a promising new target for improving the effectiveness of anti-PD-1/PD-L1 therapies [8].
曹雪涛院士最新Immunity论文:阻断癌细胞巨胞饮作用,增强癌症免疫治疗
生物世界· 2025-08-15 00:10
Core Viewpoint - The study reveals that targeting dihydroorotate dehydrogenase (DHODH) to inhibit macropinocytosis in tumor cells is a potential method to reverse immunosuppression and improve cancer immunotherapy [8]. Group 1: Key Findings - High-throughput screening identified DHODH as essential for cancer cell macropinocytosis [6]. - DHODH maintains the O-GlcNAc glycosylation modification and membrane localization of neuropilin-1 (NRP1), promoting macropinocytosis [6]. - Macropinocytosis increases the acetylation of transcription factor CIITA, leading to the suppression of major histocompatibility complex class II (MHC II) expression, thereby facilitating immune evasion [6]. Group 2: Implications for Immunotherapy - Inhibition of DHODH in cancer cells significantly enhances immune cell infiltration and activates anti-tumor immune responses, overcoming resistance to anti-PD-1 therapy [5][6]. - High expression levels of DHODH and NRP1 in human breast and lung cancer tissues correlate with poor patient prognosis [5].
Cell Genomics:李川昀/程强团队发现,让人类大脑更智慧的新基因,也让人类更易患癌
生物世界· 2025-08-05 08:00
Core Viewpoint - The study reveals that newly evolved human de novo genes, crucial for brain development and cognitive function, may also promote cancer, leading to the development of mRNA cancer vaccines that effectively stimulate anti-tumor immune responses and significantly inhibit tumor growth [3][9]. Group 1: Research Findings - The research identified 37 de novo genes with clear evolutionary trajectories unique to humans and their close primate relatives [5][7]. - These de novo genes exhibit increased expression in tumors, with 57.1% of their deletions suppressing tumor cell proliferation, indicating their oncogenic roles [5][7]. - The study highlights two specific de novo genes, ELFN1-AS1 and TYMSOS, which are expressed during early human development but reactivated in tumors, suggesting their potential as therapeutic targets [5][6]. Group 2: Vaccine Development - The research team developed an mRNA vaccine targeting ELFN1-AS1 and TYMSOS, which activates anti-tumor immune responses in humanized mouse models, demonstrating significant tumor growth inhibition [6][9]. - The new antigens derived from these genes can induce specific immune responses in patient-derived immune cells, showcasing promising clinical translation potential [6][9].
Nature:癌症免疫治疗新靶点——NNMT
生物世界· 2025-07-30 05:02
Core Viewpoint - Cancer-associated fibroblasts (CAF) play a critical role in supporting tumor growth and metastasis through extracellular matrix remodeling, paracrine signaling, and immune suppression, which limits the efficacy of immune checkpoint blockade (ICB) therapies [2][3]. Group 1 - The study published by researchers from the University of Chicago in the journal Nature reveals that NNMT promotes the recruitment of myeloid-derived suppressor cells (MDSC) into tumors via CAF, leading to the formation of an immunosuppressive tumor microenvironment (TME) [4]. - The research demonstrates that NNMT is expressed in all CAF subtypes and induces H3K27me3 hypomethylation, which facilitates the secretion of complement proteins that recruit MDSC into the tumor [7]. - The team developed a potent and specific NNMT inhibitor (NNMTi) that reduced tumor burden and metastasis in various mouse tumor models by decreasing CAF-mediated MDSC recruitment and reactivating CD8+ T cell activation, thereby restoring the efficacy of ICB therapy [9]. Group 2 - Overall, the study indicates that NNMT is a key regulatory factor of immunosuppression in the tumor microenvironment and represents a promising new target for alleviating immune suppression and enhancing cancer immunotherapy effectiveness [11].
Cell:癌症免疫大数据引擎,系统性发现基于分泌蛋白的癌症新疗法,一作已回国加入香港大学
生物世界· 2025-07-30 00:30
Core Viewpoint - The article discusses the limitations of current cancer therapies based on secreted proteins and highlights a new research study that identifies potential immunotherapy targets through a cancer immunology data platform [2][3][12]. Group 1: Research Findings - The study developed a cancer immunology data platform called CIDE, which integrates 90 multi-omics datasets covering 8,575 tumor samples across 17 types of solid tumors [8][12]. - CIDE systematically identifies genes related to immunotherapy outcomes and has revealed secreted proteins such as AOAH, CR1L, COLQ, and ADAMTS7 as new immune checkpoint blockade (ICB) regulators [9][15]. - AOAH enhances immunotherapy through dual mechanisms: increasing T cell receptor (TCR) sensitivity to weak antigens and removing inhibitory lipids that suppress dendritic cell function [10][15]. Group 2: Implications for Cancer Treatment - The findings suggest that the identified secreted proteins could serve as precise targets for a new generation of immunotherapies, overcoming the limitations of traditional therapies like IL-2 and VEGF inhibitors, which have low response rates and unstable efficacy [17]. - AOAH's validation across multiple tumor models indicates its potential as a broad-spectrum immune enhancer [18]. Group 3: Future Directions - The research aims to expand the functional map of secreted proteins, particularly focusing on the 61% of previously unreported proteins related to cancer, to discover new molecules that regulate the immune microenvironment [20]. - There is a focus on targeting lipid-immune interactions based on AOAH-related lipid metabolism pathways for developing small molecule drugs or combination therapies [20]. - The study emphasizes the need for clinical translation of molecules like AOAH as biomarkers or therapeutic targets through preclinical and clinical trials [20].
中国学者一作Nature论文:自身抗体影响癌症免疫治疗效果
生物世界· 2025-07-28 04:08
Core Viewpoint - Immune checkpoint blockade therapy has significantly changed the landscape of cancer treatment, showing strong efficacy in various cancer types, but the reasons for differential responses among patients remain unclear [1][3]. Group 1: Research Findings - A recent study published in Nature reveals that autoantibodies (AAb), typically associated with autoimmune diseases, can influence the response of cancer patients to immune checkpoint blockade therapy [3][5]. - The research involved 374 cancer patients receiving immune checkpoint blockade therapy and 131 healthy controls, mapping the immune response to 6172 extracellular and secreted proteins [5]. - The study identified approximately 3000 unique autoantibody responses in cancer patients, indicating a diverse "autoantibody response group" that has not yet reached saturation [7]. Group 2: Clinical Implications - Patients with interferon-targeting antibodies have a 40-fold higher probability of responding to treatment, contrasting with COVID-19, where similar antibodies increase mortality risk by 20-200 times [7]. - The findings suggest that targeting the exoproteome with specific autoantibodies could enhance the efficacy of immune checkpoint blockade therapy, leading to the potential development of drugs that mimic beneficial autoantibodies or neutralize harmful ones [9]. - The study also highlights that anti-TL1A antibodies enhance treatment effects by preventing T-cell apoptosis in the tumor microenvironment [12]. Group 3: Future Directions - The research opens new avenues for optimizing cancer immunotherapy by leveraging the role of autoantibodies in treatment responses, providing new targets and strategies for improving patient outcomes [9].
背靠背三篇Science论文:David Baker团队中国博后利用AI从头设计TCR,加速癌症免疫治疗
生物世界· 2025-07-25 04:05
Core Viewpoint - The article discusses the advancements in cancer immunotherapy through the use of generative AI to design artificial T-cell receptors (TCRs) that can specifically bind to pMHC complexes, overcoming the limitations of natural TCRs and enabling more precise targeting of tumor antigens [4][21][23]. Group 1: Traditional TCR Methods - Traditional methods for utilizing TCR in cancer immunotherapy involve isolating T cells from patients, either through tumor-infiltrating lymphocytes (TILs) or expanding T cells from an initial T cell library, which is technically challenging and labor-intensive [3]. - Natural TCRs often have poor affinity for tumor antigens, making it difficult to achieve effective immunotherapy [3]. Group 2: Generative AI Research - On July 24, 2025, three research papers published in the journal Science demonstrated the use of generative AI to design artificial TCRs that can bind with high specificity to pMHC complexes, thus enhancing the precision of tumor antigen targeting [4][5][21]. - The research teams involved include those from the University of Washington, Technical University of Denmark, and Stanford University [5]. Group 3: Protein Design and Functionality - The David Baker/Liu Bingxu team developed a computational method to enhance the immune system's ability to recognize and destroy cells carrying less detectable disease markers by designing proteins that specifically recognize target pMHC complexes [10]. - The designed proteins were tested against 11 different pMHC targets, including fragments from HIV and cancer-related mutations, with 8 of them successfully activating immune cells [13]. - The study confirmed that the designed proteins only bind to their specific targets, achieving atomic-level precision in construction [14]. Group 4: Rapid and Scalable Design Process - The design process demonstrated high adaptability, allowing the creation of new versions of binding proteins for different tumor and viral peptide targets in less than a week [16]. - This digital approach contrasts with traditional drug development methods, significantly shortening the drug development cycle and reducing complexity, while paving the way for more personalized therapies [16]. Group 5: Future Directions and Company Formation - The lead author, Dr. Liu Bingxu, indicated plans to establish a company to translate these research findings into therapies that can benefit patients [18]. - The research highlights the potential for artificial TCRs to revolutionize diagnostic tools and immunotherapies, particularly for diseases that currently lack effective treatments [22][23].
Cancer Cell:中山大学徐瑞华团队发现,这种肠道细菌可增强癌症免疫治疗效果
生物世界· 2025-07-25 04:05
Core Viewpoint - The emergence of immunotherapy has significantly changed the landscape of cancer treatment, but resistance to immunotherapy remains a major obstacle for its broader clinical application. Recent studies indicate that gut microbiota can enhance the efficacy of immunotherapy by modulating anti-tumor immunity [2]. Group 1: Research Findings - A study published by Professor Xu Ruihua's team from Sun Yat-sen University on July 24, 2025, in the journal Cancer Cell, demonstrates that the gut bacterium Alistipes finegoldii can enhance the efficacy of immunotherapy against solid tumors [3][4]. - The research found that a higher abundance of Alistipes finegoldii is associated with improved responses to immunotherapy, particularly enhancing the efficacy of anti-PD-1 monoclonal antibodies in solid tumor models [8]. - Alistipes finegoldii activates the CXCL16-CXCR6 signaling axis to boost anti-tumor immune responses, with lipoproteins derived from Alistipes finegoldii triggering the TLR2-NF-κB-CXCL16 signaling pathway [7][8]. Group 2: Mechanism of Action - The mechanism involves lipoproteins from Alistipes finegoldii binding to Toll-like receptor 2 (TLR2), activating the NF-κB signaling pathway, which enhances the expression of CXCL16 in CCR7+ conventional dendritic cells [7]. - The released CXCL16 aids in recruiting CXCR6+ CD8+ T cells to the tumor microenvironment (TME), effectively inhibiting tumor growth [7][8]. Group 3: Implications for Treatment - Overall, the findings suggest that combining Alistipes finegoldii with immunotherapy could represent a new strategy for treating solid tumors [10].
日本研究发现可增强癌症免疫药效果的肠道细菌
日经中文网· 2025-07-16 03:16
Core Viewpoint - The article discusses a recent Japanese study that identifies gut bacteria capable of enhancing the effectiveness of cancer immunotherapy, highlighting the potential for microbiome research in cancer treatment [1]. Group 1: Study Findings - The research indicates that specific gut bacteria can improve the response to cancer immunotherapy, suggesting a link between gut health and treatment efficacy [1]. - The study emphasizes the importance of the microbiome in influencing immune responses, which could lead to more personalized cancer treatment strategies [1]. Group 2: Implications for Cancer Treatment - The findings may pave the way for new therapeutic approaches that incorporate gut microbiota management alongside traditional cancer treatments [1]. - This research could lead to the development of probiotics or dietary interventions aimed at optimizing gut bacteria to enhance immunotherapy outcomes [1].