Workflow
癌症免疫治疗
icon
Search documents
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].
Nature:华人团队发现癌症免疫治疗新靶点——PPP2R1A
生物世界· 2025-07-06 02:50
Core Viewpoint - Immune checkpoint inhibitors (ICIs) have transformed the treatment of various solid tumors, but resistance remains a significant challenge, particularly in advanced and recurrent ovarian cancer, where response rates to single-agent PD-1/PD-L1 inhibitors are only 5%-15% [2][3] Group 1: Research Findings - A study published in Nature by a team from MD Anderson Cancer Center found that patients with PPP2R1A gene mutations had significantly improved survival after receiving combined anti-PD-1/PD-L1 and anti-CTLA-4 immunotherapy compared to those with wild-type PPP2R1A [3][6] - The presence of PPP2R1A mutations enhances tumor response to immunotherapy, and this finding was validated across various cancer types in clinical cohorts [3][9] - In recurrent ovarian cancer, dual targeting of PD-1/PD-L1 and CTLA-4 showed a response rate of 31.4% compared to 12.2% for single-agent PD-1 therapy, indicating a potential benefit for patients with ovarian clear cell carcinoma (OCCC) [5][6] Group 2: Clinical Implications - The study suggests that targeting PPP2R1A could represent an effective strategy to improve outcomes for cancer patients undergoing immunotherapy [9] - Enhanced immune cell infiltration and signaling pathways were observed in tumors with PPP2R1A mutations, indicating a more favorable immune environment for treatment [8] - The research team is conducting prospective trials to explore the efficacy of dual immune checkpoint blockade in OCCC patients, particularly those with platinum-resistant disease [5][6]
Nature Cancer:浙江大学顾臻团队等开发il-2长效缓释平台,促进癌症免疫治疗
生物世界· 2025-06-13 07:33
Core Viewpoint - The article discusses the development of a long-acting IL-2 release platform using pressure-fused biomineral tablets, which enhances antitumor immune response and addresses the limitations of traditional IL-2 therapies [3][4][10]. Group 1: Long-acting Drug Delivery - Long-acting formulations can maintain drug release for weeks, months, or even years, improving patient compliance and therapeutic efficacy [2]. - Biominers like calcium carbonate (CaC) and calcium phosphate (CaP) are promising materials for constructing long-acting formulations due to their high biocompatibility and stability [2][6]. Group 2: Research Development - The research team developed a dynamic control platform for IL-2 release through the fusion of amorphous CaC and CaP under high pressure (2 GPa) [6][7]. - A hybrid biomineral with the formula Ca(CO3)x(PO4)2(1−x)/3 was created, demonstrating crystallization-driven release behavior to optimize IL-2's in vivo fate [7]. Group 3: Experimental Results - The formulation consisted of 7.5 mg of CaC nanoparticles, 2.5 mg of CaP nanoparticles, and 30 µg of IL-2, which were completely fused under pressure to form a transparent IL-2@Ca(CO3)1/2(PO4)1/3 tablet [8]. - The Ca(CO3)1/2(PO4)1/3 tablet reshaped the immunosuppressive tumor microenvironment, enhancing the distribution of IL-2 and preferentially activating cytotoxic T cells and memory T cells, leading to weeks of IL-2 retention [9]. Group 4: Antitumor Efficacy - In a melanoma model in female mice, the Ca(CO3)1/2(PO4)1/3 tablet exhibited excellent antitumor effects, inhibiting local tumor recurrence and preventing the growth of untreated distal tumors while maintaining a long-term T cell response [10].
Immunity:细菌抗肿瘤免疫疗法有益和有害作用的细胞机制
生物世界· 2025-06-06 03:18
Core Viewpoint - The article discusses the cellular mechanisms underlying the beneficial and detrimental effects of bacterial antitumor immunotherapy, highlighting the importance of injection timing in maximizing therapeutic efficacy and minimizing tumor promotion risks [3][10]. Group 1: Research Findings - The study utilized a non-tumor antigen expressing attenuated strain of Listeria (ΔactA, Lm) to explore immune responses in tumor-bearing mice after different injection methods [5]. - Intratumoral injection (i.t.) of Lm alone recruits neutrophils that convert to an immunosuppressive phenotype, creating an immune escape microenvironment that promotes tumor growth [6]. - Conversely, intravenous injection (i.v.) induces the production of anti-Lm cytotoxic CD8+ T cells, which infiltrate the tumor upon subsequent intratumoral injection, leading to tumor suppression through apoptosis induction and enhanced antigen presentation [7][8]. Group 2: Implications of Injection Timing - The study emphasizes the significance of injection timing, suggesting that prior intravenous injection activates systemic T cells, establishing an immune foundation for subsequent intratumoral injection, thus avoiding the immunosuppressive effects associated with intratumoral injection alone [10].
开启免疫治疗新时代:华西医院仝爱平团队系统解读癌症mRNA疫苗临床应用进展及挑战
生物世界· 2025-05-22 10:04
Core Viewpoint - Cancer mRNA vaccines represent a promising new direction in cancer treatment, leveraging advancements in precision medicine and bioinformatics to enhance immune responses against tumors while addressing challenges such as tumor heterogeneity and antigen selection complexity [2][3]. Group 1: Clinical Progress and Challenges - The review summarizes the clinical progress and challenges of cancer mRNA vaccines, highlighting their ability to significantly reduce recurrence rates and induce long-term immune memory in trials for melanoma and head and neck cancers [3]. - Major obstacles include tumor heterogeneity, complex antigen selection, and vaccine stability, necessitating the integration of AI for personalized design and combination with immune checkpoint inhibitors [3][19]. Group 2: Preclinical Optimization and Development - Cancer mRNA vaccines have notable advantages, including mature synthesis technology that can encode full-length tumor antigens, enhancing T cell response efficiency without the mutation risks associated with DNA vaccines [6][8]. - Optimization efforts focus on mRNA structure (e.g., 5' cap and Poly(A) tail modifications) and delivery vehicles, with lipid nanoparticles (LNPs) being the most widely used despite challenges like hepatotoxicity [8][10]. Group 3: Mechanism of Action - The mechanism of action for mRNA cancer vaccines involves the activation of innate immune responses, where pattern recognition receptors (PRRs) identify mRNA as foreign, leading to enhanced T cell activation and immune responses against tumor cells [13][15]. Group 4: Clinical Trials and Efficacy - Ongoing clinical trials are exploring various cancer types, with mRNA vaccines encoding immune stimulators showing promise in enhancing T cell responses and improving survival rates in melanoma patients [16][18]. - Personalized neoantigen mRNA vaccines, such as Moderna's mRNA-4157, are being evaluated for their ability to significantly reduce recurrence or mortality risks in melanoma patients [17]. Group 5: Future Directions - The future of mRNA cancer vaccines is bright, with potential innovations including CARs and TCRs encoding vaccines, circRNA vaccines for sustained antigen production, and AI applications for personalized vaccine development [22][24]. - Research should focus on selecting personalized antigens, developing various immune adjuvants, and exploring the synergistic effects with gut microbiota modulation [22].
基因编辑治疗晚期胃肠道癌显成效
Ke Ji Ri Bao· 2025-05-06 23:27
Core Insights - The research team from the University of Minnesota has achieved a milestone in the treatment of advanced gastrointestinal cancers using CRISPR/Cas9 gene editing technology, confirming the safety and potential efficacy of this therapy [1] Group 1: Clinical Trial Results - The clinical trial involved 12 patients with advanced metastatic cancer, demonstrating good safety with no severe adverse reactions reported [1] - Some patients showed effective disease control, with one patient experiencing complete disappearance of metastatic tumors after several months and maintaining two years without recurrence [1] Group 2: Mechanism and Innovation - The new therapy involves genetic modification of tumor-infiltrating lymphocytes (TILs) to inactivate the CISH gene, allowing for more precise identification and attack on cancer cells [1] - Unlike traditional cancer therapies that require repeated dosing, this gene editing therapy can achieve lasting effects through a one-time modification of T cells [1] Group 3: Future Directions - The research team successfully cultivated and infused over 10 billion engineered TIL cells, validating the feasibility of large-scale clinical-grade cell preparation [2] - Despite promising initial results, the existing processes face challenges such as high costs and complexity, prompting plans to optimize treatment protocols and explore mechanisms of efficacy differences [2]
Nature Cancer:厦门大学刘文/夏宁邵/罗文新团队发现新的癌症治疗免疫检查点——PILRα
生物世界· 2025-05-05 05:40
Core Viewpoint - The study identifies PILRα as a potential immune checkpoint in cancer immunotherapy, which interacts with T cell surface protein CD99 to suppress anti-tumor immunity, indicating its clinical significance in various human cancers with poor prognosis [2][4][5]. Group 1 - The research published in Nature Cancer reveals that PILRα expressed on tumor cells inhibits T cell activation, proliferation, and effector functions by targeting CD99, affecting the ZAP70/NFAT/IL-2/JAK/STAT signaling pathway [3]. - Blocking the interaction between PILRα and CD99 using specific antibodies significantly enhances T cell anti-tumor immune responses and suppresses tumor growth, showing synergistic effects when combined with anti-PD-1 antibodies [3][5]. - High expression of PILRα in various human cancers is associated with unfavorable prognosis, highlighting its potential as a therapeutic target in cancer treatment [4].