首页 > 最新文献

Cancer immunology research最新文献

英文 中文
Multifunctional Bispecific Nanovesicles Targeting SLAMF7 Trigger Potent Antitumor Immunity. 以 SLAMF7 为靶点的多功能双特异性纳米囊泡可触发强效抗肿瘤免疫。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-01 DOI: 10.1158/2326-6066.CIR-23-1102
Manman Zhu, Yongjian Wu, Tianchuan Zhu, Jian Chen, Zhenxing Chen, Hanxi Ding, Siyi Tan, Jianzhong He, Qi Zeng, Xi Huang

The effectiveness of immune checkpoint inhibitor (ICI) therapy is hindered by the ineffective infiltration and functioning of cytotoxic T cells and the immunosuppressive tumor microenvironment (TME). Signaling lymphocytic activation molecule family member 7 (SLAMF7) is a pivotal co-stimulatory receptor thought to simultaneously trigger NK-cell, T-cell, and macrophage antitumor cytotoxicity. However, the potential of this collaborative immune stimulation in antitumor immunity for solid tumors is underexplored due to the exclusive expression of SLAMF7 by hematopoietic cells. Here, we report the development and characterization of multifunctional bispecific nanovesicles (NVs) targeting SLAMF7 and glypican-3-a hepatocellular carcinoma (HCC)-specific tumor antigen. We found that by effectively "decorating" the surfaces of solid tumors with SLAMF7, these NVs directly induced potent and specific antitumor immunity and remodeled the immunosuppressive TME, sensitizing the tumors to programmed cell death protein 1 (PD1) blockade. Our findings highlight the potential of SLAMF7-targeted multifunctional bispecific NVs as an anticancer strategy with implications for designing next-generation targeted cancer therapies.

免疫检查点抑制剂(ICI)疗法的有效性因细胞毒性 T 细胞的无效浸润和运作以及免疫抑制性肿瘤微环境(TME)而受到阻碍。信号淋巴细胞活化分子家族成员 7(SLAMF7)是一种关键的协同刺激受体,被认为能同时触发自然杀伤(NK)细胞、T 细胞和巨噬细胞的抗肿瘤细胞毒性。然而,由于 SLAMF7 仅由造血细胞表达,这种协同免疫刺激在实体瘤抗肿瘤免疫中的潜力还未得到充分探索。在此,我们报告了靶向 SLAMF7 和 Glypican-3(肝细胞癌(HCC)特异性肿瘤抗原)的多功能双特异性纳米囊泡的开发和表征。我们发现,通过用 SLAMF7 有效地 "装饰 "实体瘤表面,这些纳米颗粒可直接诱导强效特异性抗肿瘤免疫,并重塑免疫抑制性 TME,使肿瘤对程序性细胞死亡蛋白 1 (PD-1) 阻滞剂敏感。我们的研究结果凸显了SLAMF7靶向多功能双特异性纳米颗粒作为一种抗癌策略的潜力,对设计下一代癌症靶向疗法具有重要意义。
{"title":"Multifunctional Bispecific Nanovesicles Targeting SLAMF7 Trigger Potent Antitumor Immunity.","authors":"Manman Zhu, Yongjian Wu, Tianchuan Zhu, Jian Chen, Zhenxing Chen, Hanxi Ding, Siyi Tan, Jianzhong He, Qi Zeng, Xi Huang","doi":"10.1158/2326-6066.CIR-23-1102","DOIUrl":"10.1158/2326-6066.CIR-23-1102","url":null,"abstract":"<p><p>The effectiveness of immune checkpoint inhibitor (ICI) therapy is hindered by the ineffective infiltration and functioning of cytotoxic T cells and the immunosuppressive tumor microenvironment (TME). Signaling lymphocytic activation molecule family member 7 (SLAMF7) is a pivotal co-stimulatory receptor thought to simultaneously trigger NK-cell, T-cell, and macrophage antitumor cytotoxicity. However, the potential of this collaborative immune stimulation in antitumor immunity for solid tumors is underexplored due to the exclusive expression of SLAMF7 by hematopoietic cells. Here, we report the development and characterization of multifunctional bispecific nanovesicles (NVs) targeting SLAMF7 and glypican-3-a hepatocellular carcinoma (HCC)-specific tumor antigen. We found that by effectively \"decorating\" the surfaces of solid tumors with SLAMF7, these NVs directly induced potent and specific antitumor immunity and remodeled the immunosuppressive TME, sensitizing the tumors to programmed cell death protein 1 (PD1) blockade. Our findings highlight the potential of SLAMF7-targeted multifunctional bispecific NVs as an anticancer strategy with implications for designing next-generation targeted cancer therapies.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1007-1021"},"PeriodicalIF":8.1,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141178296","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Impact of scFv on Functionality and Safety of Third-Generation CD123 CAR T Cells. scFv 对第三代 CD123 CAR T 细胞功能性和安全性的影响。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-01 DOI: 10.1158/2326-6066.CIR-23-0548
Maxime Fredon, Margaux Poussard, Sabeha Biichlé, Francis Bonnefoy, Charles-Frédéric Mantion, Evan Seffar, Florian Renosi, Elodie Bôle-Richard, Romain Boidot, Sandrine Chevrier, François Anna, Maria Loustau, Julien Caumartin, Mathieu Gonçalves-Venturelli, Eric Robinet, Philippe Saas, Eric Deconinck, Etienne Daguidau, Xavier Roussel, Yann Godet, Olivier Adotévi, Fanny Angelot-Delettre, Jeanne Galaine, Francine Garnache-Ottou

Chimeric antigen receptor (CAR) T cells express an extracellular domain consisting of a single-chain fragment variable (scFv) targeting a surface tumor-associated antigen. scFv selection should involve safety profiling with evaluation of the efficacy/toxicity balance, especially when the target antigen also is expressed on healthy cells. Here, to assess differences in terms of efficacy and on-target/off-tumor effects, we generated five different CARs targeting CD123 by substituting only the scFv. In in vitro models, T cells engineered to express three of these five CD123 CARs were effectively cytotoxic on leukemic cells without increasing lysis of monocytes or endothelial cells. Using the IncuCyte system, we confirmed the low cytotoxicity of CD123 CAR T cells on endothelial cells. Hematotoxicity evaluation using progenitor culture and CD34 cell lysis showed that two of the five CD123 CAR T cells were less cytotoxic on hematopoietic stem cells. Using a humanized mouse model, we confirmed that CD123- cells were not eliminated by the CD123 CAR T cells. Two CD123 CAR T cells reduced tumor infiltration and increased the overall survival of mice in three in vivo models of blastic plasmacytoid dendritic cell neoplasm. In an aggressive version of this model, bulk RNA sequencing analysis showed that these CD123 CAR T cells upregulated genes associated with cytotoxicity and activation/exhaustion a few days after the injection. Together, these results emphasize the importance of screening different scFvs for the development of CAR constructs to support selection of cells with the optimal risk-benefit ratio for clinical development.

嵌合抗原受体(CAR)T细胞表达由单链片段变量(scFv)组成的胞外结构域,靶向表面肿瘤相关抗原。scFv的选择应包括安全性分析和疗效/毒性平衡评估,尤其是当靶抗原也在健康细胞中表达时。在这里,为了评估疗效和靶向/非肿瘤效应方面的差异,我们仅用 scFv 替代了五种不同的 CD123 靶向 CAR。在体外模型中,表达这五种 CD123 CARs 中三种的 T 细胞对白血病细胞具有有效的细胞毒性,同时不会增加对单核细胞或内皮细胞的裂解。我们使用 IncuCyte® 系统证实了 CD123 CAR T 细胞对内皮细胞的低细胞毒性。利用祖细胞培养和 CD34 细胞裂解进行的血液毒性评估显示,五种 CD123 CAR T 细胞中有两种对造血干细胞的细胞毒性较低。通过人源化小鼠模型,我们证实 CD123 CAR T 细胞不会消灭 CD123 细胞。在三种疱性浆细胞树突状细胞瘤体内模型中,两种CD123 CAR T细胞减少了肿瘤浸润,提高了小鼠的总体存活率。在该模型的侵袭性版本中,大量 RNA 测序分析表明,这些 CD123 CAR T 细胞在注射几天后上调了与细胞毒性和活化/衰竭相关的基因。这些结果共同强调了在开发 CAR 构建物时筛选不同 scFvs 的重要性,以便为临床开发选择具有最佳风险效益比的细胞。
{"title":"Impact of scFv on Functionality and Safety of Third-Generation CD123 CAR T Cells.","authors":"Maxime Fredon, Margaux Poussard, Sabeha Biichlé, Francis Bonnefoy, Charles-Frédéric Mantion, Evan Seffar, Florian Renosi, Elodie Bôle-Richard, Romain Boidot, Sandrine Chevrier, François Anna, Maria Loustau, Julien Caumartin, Mathieu Gonçalves-Venturelli, Eric Robinet, Philippe Saas, Eric Deconinck, Etienne Daguidau, Xavier Roussel, Yann Godet, Olivier Adotévi, Fanny Angelot-Delettre, Jeanne Galaine, Francine Garnache-Ottou","doi":"10.1158/2326-6066.CIR-23-0548","DOIUrl":"10.1158/2326-6066.CIR-23-0548","url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR) T cells express an extracellular domain consisting of a single-chain fragment variable (scFv) targeting a surface tumor-associated antigen. scFv selection should involve safety profiling with evaluation of the efficacy/toxicity balance, especially when the target antigen also is expressed on healthy cells. Here, to assess differences in terms of efficacy and on-target/off-tumor effects, we generated five different CARs targeting CD123 by substituting only the scFv. In in vitro models, T cells engineered to express three of these five CD123 CARs were effectively cytotoxic on leukemic cells without increasing lysis of monocytes or endothelial cells. Using the IncuCyte system, we confirmed the low cytotoxicity of CD123 CAR T cells on endothelial cells. Hematotoxicity evaluation using progenitor culture and CD34 cell lysis showed that two of the five CD123 CAR T cells were less cytotoxic on hematopoietic stem cells. Using a humanized mouse model, we confirmed that CD123- cells were not eliminated by the CD123 CAR T cells. Two CD123 CAR T cells reduced tumor infiltration and increased the overall survival of mice in three in vivo models of blastic plasmacytoid dendritic cell neoplasm. In an aggressive version of this model, bulk RNA sequencing analysis showed that these CD123 CAR T cells upregulated genes associated with cytotoxicity and activation/exhaustion a few days after the injection. Together, these results emphasize the importance of screening different scFvs for the development of CAR constructs to support selection of cells with the optimal risk-benefit ratio for clinical development.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1090-1107"},"PeriodicalIF":8.1,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141178316","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Overexpression of an Engineered SERPINB9 Enhances Allogeneic T-cell Persistence and Efficacy. 过表达工程化 SerpinB9 可增强异体 T 细胞的存活率和疗效
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-01 DOI: 10.1158/2326-6066.CIR-23-1001
Pei Y Teo, Youngrock Jung, David H Quach, Joanna Koh, Richard W Ong, Angeline Goh, Alrina Tan, Chee H Ng, Cheah C Seh, Kar W Tan, Ivan D Horak, Lionel Low

Allogeneic chimeric antigen receptor (CAR)-expressing T cells offer many advantages over autologous therapies, but their benefits are curtailed by graft-versus-host disease and elimination by recipient immune cells. Moreover, just as with autologous therapies, allogeneic CAR T cells are susceptible to activation-induced cell death (AICD) caused by chronic antigen exposure (CAE). Granzyme B- and Fas/Fas ligand-initiated caspase-mediated apoptoses are key mechanisms of T-cell death caused by T/NK cell-mediated allorejection or CAE. We explored a protective strategy of engineering CAR T cells to overexpress variants of the Granzyme B-specific serine protease inhibitor SERPINB9 (SB9) to improve allogeneic T-cell persistence and antitumor efficacy. We showed that the overexpression of an SB9 variant with broadened caspase specificity, SB9(CAS), not only significantly reduced rejection of allogeneic CAR T cells but also increased their resistance to AICD and enabled them to thrive better under CAE, thus improving allogeneic T-cell persistence and antitumor activity in vitro and in vivo. In addition, although SB9(CAS) overexpression improved the efficacy of allogeneic CAR T-cell therapy by conferring protection to cell death, we did not observe any autonomous growth, and the engineered CAR T cells were still susceptible to an inducible suicide switch. Hence, SB9(CAS) overexpression is a promising strategy that can strengthen current development of cell therapies, broadening their applications to address unmet medical needs.

与自体疗法相比,表达嵌合抗原受体(CAR)的异体 T 细胞具有许多优势,但它们的益处因移植物抗宿主疾病(GvHD)和受体免疫细胞的清除而受到限制。此外,与自体疗法一样,异体 CAR T 细胞也容易受到慢性抗原暴露(CAE)引起的活化诱导细胞死亡(AICD)的影响。颗粒酶 B(GzmB)和 Fas/FasL 引发、Caspase 介导的细胞凋亡是 T/NK 细胞介导的异体排斥或 CAE 导致 T 细胞死亡的关键机制。我们探索了一种保护性策略,即设计 CAR T 细胞,使其过度表达 GzmB 特异性丝氨酸蛋白酶抑制剂 SerpinB9(SB9)的变体,以提高异体 T 细胞的持久性和抗肿瘤疗效。我们的研究表明,过表达一种具有更广泛的caspase特异性的SB9变体SB9(CAS),不仅能显著降低异体CAR T细胞的排斥反应,还能增强它们对AICD的抵抗力,使它们在CAE下更好地生长,从而提高异体T细胞在体外和体内的持久性和抗肿瘤活性。此外,虽然SB9(CAS)外表达能保护细胞不死亡,从而提高异基因CAR T细胞疗法的疗效,但我们并没有观察到任何自主生长,而且工程化的CAR T细胞仍然易受诱导性自杀开关的影响。因此,SB9(CAS)外表达是一种很有前景的策略,它可以加强目前细胞疗法的发展,扩大其应用范围,以满足未得到满足的医疗需求。
{"title":"Overexpression of an Engineered SERPINB9 Enhances Allogeneic T-cell Persistence and Efficacy.","authors":"Pei Y Teo, Youngrock Jung, David H Quach, Joanna Koh, Richard W Ong, Angeline Goh, Alrina Tan, Chee H Ng, Cheah C Seh, Kar W Tan, Ivan D Horak, Lionel Low","doi":"10.1158/2326-6066.CIR-23-1001","DOIUrl":"10.1158/2326-6066.CIR-23-1001","url":null,"abstract":"<p><p>Allogeneic chimeric antigen receptor (CAR)-expressing T cells offer many advantages over autologous therapies, but their benefits are curtailed by graft-versus-host disease and elimination by recipient immune cells. Moreover, just as with autologous therapies, allogeneic CAR T cells are susceptible to activation-induced cell death (AICD) caused by chronic antigen exposure (CAE). Granzyme B- and Fas/Fas ligand-initiated caspase-mediated apoptoses are key mechanisms of T-cell death caused by T/NK cell-mediated allorejection or CAE. We explored a protective strategy of engineering CAR T cells to overexpress variants of the Granzyme B-specific serine protease inhibitor SERPINB9 (SB9) to improve allogeneic T-cell persistence and antitumor efficacy. We showed that the overexpression of an SB9 variant with broadened caspase specificity, SB9(CAS), not only significantly reduced rejection of allogeneic CAR T cells but also increased their resistance to AICD and enabled them to thrive better under CAE, thus improving allogeneic T-cell persistence and antitumor activity in vitro and in vivo. In addition, although SB9(CAS) overexpression improved the efficacy of allogeneic CAR T-cell therapy by conferring protection to cell death, we did not observe any autonomous growth, and the engineered CAR T cells were still susceptible to an inducible suicide switch. Hence, SB9(CAS) overexpression is a promising strategy that can strengthen current development of cell therapies, broadening their applications to address unmet medical needs.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1108-1122"},"PeriodicalIF":8.1,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141236953","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Sampling of Highlights from the Literature. 文献精华选集》。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-02 DOI: 10.1158/2326-6066.CIR-12-7-WWR
{"title":"A Sampling of Highlights from the Literature.","authors":"","doi":"10.1158/2326-6066.CIR-12-7-WWR","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-12-7-WWR","url":null,"abstract":"","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":"12 7","pages":"797"},"PeriodicalIF":8.1,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141475980","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IL3-Driven T Cell-Basophil Crosstalk Enhances Antitumor Immunity. IL-3 驱动的 T 细胞-嗜碱性粒细胞串联增强了抗肿瘤免疫力。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-02 DOI: 10.1158/2326-6066.CIR-23-0851
Jian Wei, Colleen L Mayberry, Xiaoting Lv, Fangyan Hu, Taushif Khan, Natalie A Logan, John J Wilson, John D Sears, Damien Chaussabel, Chih-Hao Chang

Cytotoxic T lymphocytes (CTL) are pivotal in combating cancer, yet their efficacy is often hindered by the immunosuppressive tumor microenvironment, resulting in CTL exhaustion. This study investigates the role of interleukin-3 (IL3) in orchestrating antitumor immunity through CTL modulation. We found that intratumoral CTLs exhibited a progressive decline in IL3 production, which was correlated with impaired cytotoxic function. Augmenting IL3 supplementation, through intraperitoneal administration of recombinant IL3, IL3-expressing tumor cells, or IL3-engineered CD8+ T cells, conferred protection against tumor progression, concomitant with increased CTL activity. CTLs were critical for this therapeutic efficacy as IL3 demonstrated no impact on tumor growth in Rag1 knockout mice or following CD8+ T-cell depletion. Rather than acting directly, CTL-derived IL3 exerted its influence on basophils, concomitantly amplifying antitumor immunity within CTLs. Introducing IL3-activated basophils retarded tumor progression, whereas basophil depletion diminished the effectiveness of IL3 supplementation. Furthermore, IL3 prompted basophils to produce IL4, which subsequently elevated CTL IFNγ production and viability. Further, the importance of basophil-derived IL4 was evident from the absence of benefits of IL3 supplementation in IL4 knockout tumor-bearing mice. Overall, this research has unveiled a role for IL3-mediated CTL-basophil cross-talk in regulating antitumor immunity and suggests harnessing IL3 sustenance as a promising approach for optimizing and enhancing cancer immunotherapy. See related Spotlight, p. 798.

细胞毒性T淋巴细胞(CTL)在抗癌过程中发挥着关键作用,但它们的功效往往受到免疫抑制性肿瘤微环境的阻碍,从而导致衰竭。本研究探讨了白细胞介素(IL)-3 在通过 CTL 调节抗肿瘤免疫中的作用。瘤内 CTL 的 IL-3 产量会逐渐下降,这与细胞毒性功能受损有关。通过腹腔注射重组IL-3、表达IL-3的肿瘤细胞或IL-3设计的CD8+T细胞来增强IL-3的补充,可在提高CTL活性的同时保护患者免受肿瘤进展的影响。CTL 对这种疗效至关重要,因为 IL-3 对 Rag1 基因敲除小鼠或 CD8+ T 细胞耗竭后的肿瘤生长没有影响。CTL衍生的IL-3不是直接发挥作用,而是对嗜碱性粒细胞施加影响,协同放大CTL内的抗肿瘤免疫力。引入 IL-3 激活的嗜碱性粒细胞可延缓肿瘤进展,而嗜碱性粒细胞耗竭则会降低补充 IL-3 的效果。此外,IL-3 还能促使嗜碱性粒细胞产生 IL-4,从而提高 CTL IFN-γ 的产生和活力。值得注意的是,嗜碱性粒细胞产生的IL-4的重要性从IL-4基因敲除的肿瘤小鼠缺乏IL-3补充剂的益处中可见一斑。总之,这项研究揭示了IL-3介导的CTL-嗜碱性粒细胞在调节抗肿瘤免疫中的串联作用,并为利用IL-3维持作为优化和增强癌症免疫疗法的一种有前途的方法提供了前景。
{"title":"IL3-Driven T Cell-Basophil Crosstalk Enhances Antitumor Immunity.","authors":"Jian Wei, Colleen L Mayberry, Xiaoting Lv, Fangyan Hu, Taushif Khan, Natalie A Logan, John J Wilson, John D Sears, Damien Chaussabel, Chih-Hao Chang","doi":"10.1158/2326-6066.CIR-23-0851","DOIUrl":"10.1158/2326-6066.CIR-23-0851","url":null,"abstract":"<p><p>Cytotoxic T lymphocytes (CTL) are pivotal in combating cancer, yet their efficacy is often hindered by the immunosuppressive tumor microenvironment, resulting in CTL exhaustion. This study investigates the role of interleukin-3 (IL3) in orchestrating antitumor immunity through CTL modulation. We found that intratumoral CTLs exhibited a progressive decline in IL3 production, which was correlated with impaired cytotoxic function. Augmenting IL3 supplementation, through intraperitoneal administration of recombinant IL3, IL3-expressing tumor cells, or IL3-engineered CD8+ T cells, conferred protection against tumor progression, concomitant with increased CTL activity. CTLs were critical for this therapeutic efficacy as IL3 demonstrated no impact on tumor growth in Rag1 knockout mice or following CD8+ T-cell depletion. Rather than acting directly, CTL-derived IL3 exerted its influence on basophils, concomitantly amplifying antitumor immunity within CTLs. Introducing IL3-activated basophils retarded tumor progression, whereas basophil depletion diminished the effectiveness of IL3 supplementation. Furthermore, IL3 prompted basophils to produce IL4, which subsequently elevated CTL IFNγ production and viability. Further, the importance of basophil-derived IL4 was evident from the absence of benefits of IL3 supplementation in IL4 knockout tumor-bearing mice. Overall, this research has unveiled a role for IL3-mediated CTL-basophil cross-talk in regulating antitumor immunity and suggests harnessing IL3 sustenance as a promising approach for optimizing and enhancing cancer immunotherapy. See related Spotlight, p. 798.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"822-839"},"PeriodicalIF":8.1,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11219266/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140911428","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Do Not Forget the Granulocytic Compartment's Role in T cell-Mediated Antitumor Immunity. 不要忘记粒细胞区在 T 细胞介导的抗肿瘤免疫中的作用。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-02 DOI: 10.1158/2326-6066.CIR-24-0395
Udo S Gaipl

Antitumor immune responses are predominantly mediated by CD8+ cytotoxic T cells (CTLs). But immune-modulatory factors in the tumor microenvironment determine the effectiveness of these responses. In this issue, Wei and colleagues report a new role for CTL-derived IL3 in stimulating basophilic granulocytes to produce IL4, which, in turn, activates, reprograms, and stabilizes CTLs. These findings stress the importance of the crosstalk between the innate and adaptive immune systems to elicit efficient antitumor immunity. See related article by Wei et al., p. 822 (3).

抗肿瘤免疫反应主要由 CD8+ 细胞毒性 T 细胞(CTL)介导。但肿瘤微环境中的免疫调节因子决定了这些反应的有效性。在本期杂志中,Wei及其同事报告了CTL衍生的IL3在刺激嗜碱性粒细胞产生IL4方面的新作用,而IL4反过来又能激活、重编程和稳定CTL。这些发现强调了先天性免疫系统和适应性免疫系统之间的相互作用对激发高效抗肿瘤免疫的重要性。参见 Wei 等人的相关文章,第 822 页(3)。
{"title":"Do Not Forget the Granulocytic Compartment's Role in T cell-Mediated Antitumor Immunity.","authors":"Udo S Gaipl","doi":"10.1158/2326-6066.CIR-24-0395","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-24-0395","url":null,"abstract":"<p><p>Antitumor immune responses are predominantly mediated by CD8+ cytotoxic T cells (CTLs). But immune-modulatory factors in the tumor microenvironment determine the effectiveness of these responses. In this issue, Wei and colleagues report a new role for CTL-derived IL3 in stimulating basophilic granulocytes to produce IL4, which, in turn, activates, reprograms, and stabilizes CTLs. These findings stress the importance of the crosstalk between the innate and adaptive immune systems to elicit efficient antitumor immunity. See related article by Wei et al., p. 822 (3).</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":"12 7","pages":"798-799"},"PeriodicalIF":8.1,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141475981","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PVRIG is Expressed on Stem-Like T Cells in Dendritic Cell-Rich Niches in Tumors and Its Blockade May Induce Immune Infiltration in Non-Inflamed Tumors. PVRIG 在肿瘤中树突状细胞丰富的龛位中的干样 T 细胞上表达,阻断 PVRIG 可诱导非炎症肿瘤中的免疫浸润。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-07-02 DOI: 10.1158/2326-6066.CIR-23-0752
Zoya Alteber, Gady Cojocaru, Roy Z Granit, Inbal Barbiro, Assaf Wool, Masha Frenkel, Amit Novik, Adi Shuchami, Yu Liang, Vered D Carmi, Niv Sabath, Rob Foreman, Natalia Petrenko, Jiang He, Yossef Kliger, Adva Levy-Barda, Ram Eitan, Oded Raban, Eran Sadot, Omri Sulimani, Abraham Avi Nathan, Henry Adewoye, Pierre Ferre, Zurit Levine, Eran Ophir

Cancers that are poorly immune infiltrated pose a substantial challenge, with current immunotherapies yielding limited clinical success. Stem-like memory T cells (TSCM) have been identified as a subgroup of T cells that possess strong proliferative capacity and that can expand and differentiate following interactions with dendritic cells (DCs). In this study, we explored the pattern of expression of a recently discovered inhibitory receptor poliovirus receptor-related immunoglobulin domain protein (PVRIG) and its ligand, poliovirus receptor-related ligand 2 (PVRL2), in the human tumor microenvironment. Using spatial and single-cell RNA transcriptomics data across diverse cancer indications, we found that among the T-cell checkpoints, PVRIG is uniquely expressed on TSCM and PVRL2 is expressed on DCs in immune aggregate niches in tumors. PVRIG blockade could therefore enhance TSCM-DC interactions and efficiently drive T-cell infiltration to tumors. Consistent with these data, following PVRIG blockade in patients with poorly infiltrated tumors, we observed immune modulation including increased tumor T-cell infiltration, T-cell receptor (TCR) clonality, and intratumoral T-cell expansion, all of which were associated with clinical benefit. These data suggest PVRIG blockade as a promising strategy to induce potent antitumor T-cell responses, providing a novel approach to overcome resistance to immunotherapy in immune-excluded tumors.

免疫浸润较差的癌症是一个巨大的挑战,目前的免疫疗法在临床上收效甚微。干样记忆 T 细胞(TSCM)已被确定为 T 细胞的一个亚群,它具有很强的增殖能力,并能在与树突状细胞(DC)相互作用后扩增和分化。在这项研究中,我们探讨了最近发现的抑制性受体 PVRIG 及其配体 PVRL2 在人类肿瘤微环境中的表达模式。利用不同癌症适应症的空间和单细胞 RNA 转录组学数据,我们发现在 T 细胞检查点中,PVRIG 在 TSCM 上有独特的表达,而 PVRL2 则在肿瘤免疫聚集龛中的 DC 上表达。因此,阻断 PVRIG 可增强 TSCM-DC 相互作用,并有效推动 T 细胞向肿瘤浸润。与这些数据相一致的是,在对浸润较差的肿瘤患者进行 PVRIG 阻断后,我们观察到了免疫调节,包括肿瘤 T 细胞浸润增加、T 细胞受体 (TCR) 克隆和瘤内 T 细胞扩增,所有这些都与临床获益相关。这些数据表明,PVRIG阻断是诱导强效抗肿瘤T细胞反应的一种有前途的策略,为克服免疫排斥肿瘤对免疫疗法的耐药性提供了一种新方法。
{"title":"PVRIG is Expressed on Stem-Like T Cells in Dendritic Cell-Rich Niches in Tumors and Its Blockade May Induce Immune Infiltration in Non-Inflamed Tumors.","authors":"Zoya Alteber, Gady Cojocaru, Roy Z Granit, Inbal Barbiro, Assaf Wool, Masha Frenkel, Amit Novik, Adi Shuchami, Yu Liang, Vered D Carmi, Niv Sabath, Rob Foreman, Natalia Petrenko, Jiang He, Yossef Kliger, Adva Levy-Barda, Ram Eitan, Oded Raban, Eran Sadot, Omri Sulimani, Abraham Avi Nathan, Henry Adewoye, Pierre Ferre, Zurit Levine, Eran Ophir","doi":"10.1158/2326-6066.CIR-23-0752","DOIUrl":"10.1158/2326-6066.CIR-23-0752","url":null,"abstract":"<p><p>Cancers that are poorly immune infiltrated pose a substantial challenge, with current immunotherapies yielding limited clinical success. Stem-like memory T cells (TSCM) have been identified as a subgroup of T cells that possess strong proliferative capacity and that can expand and differentiate following interactions with dendritic cells (DCs). In this study, we explored the pattern of expression of a recently discovered inhibitory receptor poliovirus receptor-related immunoglobulin domain protein (PVRIG) and its ligand, poliovirus receptor-related ligand 2 (PVRL2), in the human tumor microenvironment. Using spatial and single-cell RNA transcriptomics data across diverse cancer indications, we found that among the T-cell checkpoints, PVRIG is uniquely expressed on TSCM and PVRL2 is expressed on DCs in immune aggregate niches in tumors. PVRIG blockade could therefore enhance TSCM-DC interactions and efficiently drive T-cell infiltration to tumors. Consistent with these data, following PVRIG blockade in patients with poorly infiltrated tumors, we observed immune modulation including increased tumor T-cell infiltration, T-cell receptor (TCR) clonality, and intratumoral T-cell expansion, all of which were associated with clinical benefit. These data suggest PVRIG blockade as a promising strategy to induce potent antitumor T-cell responses, providing a novel approach to overcome resistance to immunotherapy in immune-excluded tumors.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"876-890"},"PeriodicalIF":8.1,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140944097","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Engineering CD3/CD137 Dual Specificity into a DLL3-Targeted T-Cell Engager Enhances T-Cell Infiltration and Efficacy against Small-Cell Lung Cancer. 将 CD3/CD137 双特异性植入 DLL3 靶向 T 细胞吸引器,可增强 T 细胞浸润和对小细胞肺癌的疗效。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-23-0638
Hirofumi Mikami, Shu Feng, Yutaka Matsuda, Shinya Ishii, Sotaro Naoi, Yumiko Azuma, Hiroaki Nagano, Kentaro Asanuma, Yoko Kayukawa, Toshiaki Tsunenari, Shogo Kamikawaji, Ryutaro Iwabuchi, Junko Shinozuka, Masaki Yamazaki, Haruka Kuroi, Samantha Shu Wen Ho, Siok Wan Gan, Priyanka Chichili, Chai Ling Pang, Chiew Ying Yeo, Shun Shimizu, Naoka Hironiwa, Yasuko Kinoshita, Yuichiro Shimizu, Akihisa Sakamoto, Masaru Muraoka, Noriyuki Takahashi, Tatsuya Kawa, Hirotake Shiraiwa, Futa Mimoto, Kenji Kashima, Mika Kamata-Sakurai, Shumpei Ishikawa, Hiroyuki Aburatani, Takehisa Kitazawa, Tomoyuki Igawa

Small-cell lung cancer (SCLC) is an aggressive cancer for which immune checkpoint inhibitors (ICI) have had only limited success. Bispecific T-cell engagers are promising therapeutic alternatives for ICI-resistant tumors, but not all patients with SCLC are responsive. Herein, to integrate CD137 costimulatory function into a T-cell engager format and thereby augment therapeutic efficacy, we generated a CD3/CD137 dual-specific Fab and engineered a DLL3-targeted trispecific antibody (DLL3 trispecific). The CD3/CD137 dual-specific Fab was generated to competitively bind to CD3 and CD137 to prevent DLL3-independent cross-linking of CD3 and CD137, which could lead to systemic T-cell activation. We demonstrated that DLL3 trispecific induced better tumor growth control and a marked increase in the number of intratumoral T cells compared with a conventional DLL3-targeted bispecific T-cell engager. These findings suggest that DLL3 trispecific can exert potent efficacy by inducing concurrent CD137 costimulation and provide a promising therapeutic option for SCLC.

小细胞肺癌(SCLC)是一种侵袭性癌症,免疫检查点抑制剂(ICIs)对其治疗效果有限。双特异性T细胞吸引剂是治疗对ICI耐药的肿瘤的有希望的替代疗法,但并非所有SCLC患者都有反应。在此,为了将 CD137 的调控功能整合到 T 细胞吸引剂中,从而提高疗效,我们生成了 CD3/CD137 双特异性 Fab,并设计了 DLL3 靶向三特异性抗体(DLL3 三特异性)。生成的 CD3/CD137 双特异性 Fab 能竞争性地与 CD3 和 CD137 结合,以防止 CD3 和 CD137 在不依赖 DLL3 的情况下发生交联,从而导致全身性 T 细胞活化。我们证明,与传统的 DLL3 靶向双特异性 T 细胞吞噬剂相比,DLL3 三特异性能更好地控制肿瘤生长,并显著增加瘤内 T 细胞的数量。这些研究结果表明,DLL3三特异性可通过同时诱导CD137成本刺激发挥强大的疗效,为SCLC的治疗提供了一种前景广阔的选择。
{"title":"Engineering CD3/CD137 Dual Specificity into a DLL3-Targeted T-Cell Engager Enhances T-Cell Infiltration and Efficacy against Small-Cell Lung Cancer.","authors":"Hirofumi Mikami, Shu Feng, Yutaka Matsuda, Shinya Ishii, Sotaro Naoi, Yumiko Azuma, Hiroaki Nagano, Kentaro Asanuma, Yoko Kayukawa, Toshiaki Tsunenari, Shogo Kamikawaji, Ryutaro Iwabuchi, Junko Shinozuka, Masaki Yamazaki, Haruka Kuroi, Samantha Shu Wen Ho, Siok Wan Gan, Priyanka Chichili, Chai Ling Pang, Chiew Ying Yeo, Shun Shimizu, Naoka Hironiwa, Yasuko Kinoshita, Yuichiro Shimizu, Akihisa Sakamoto, Masaru Muraoka, Noriyuki Takahashi, Tatsuya Kawa, Hirotake Shiraiwa, Futa Mimoto, Kenji Kashima, Mika Kamata-Sakurai, Shumpei Ishikawa, Hiroyuki Aburatani, Takehisa Kitazawa, Tomoyuki Igawa","doi":"10.1158/2326-6066.CIR-23-0638","DOIUrl":"10.1158/2326-6066.CIR-23-0638","url":null,"abstract":"<p><p>Small-cell lung cancer (SCLC) is an aggressive cancer for which immune checkpoint inhibitors (ICI) have had only limited success. Bispecific T-cell engagers are promising therapeutic alternatives for ICI-resistant tumors, but not all patients with SCLC are responsive. Herein, to integrate CD137 costimulatory function into a T-cell engager format and thereby augment therapeutic efficacy, we generated a CD3/CD137 dual-specific Fab and engineered a DLL3-targeted trispecific antibody (DLL3 trispecific). The CD3/CD137 dual-specific Fab was generated to competitively bind to CD3 and CD137 to prevent DLL3-independent cross-linking of CD3 and CD137, which could lead to systemic T-cell activation. We demonstrated that DLL3 trispecific induced better tumor growth control and a marked increase in the number of intratumoral T cells compared with a conventional DLL3-targeted bispecific T-cell engager. These findings suggest that DLL3 trispecific can exert potent efficacy by inducing concurrent CD137 costimulation and provide a promising therapeutic option for SCLC.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"719-730"},"PeriodicalIF":8.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140334853","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The Exonuclease TREX1 Constitutes an Innate Immune Checkpoint Limiting cGAS/STING-Mediated Antitumor Immunity. 外切酶 TREX1 是限制 cGAS/STING 介导的抗肿瘤免疫的先天免疫检查点。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-23-1078
Junghyun Lim, Ryan Rodriguez, Katherine Williams, John Silva, Alan G Gutierrez, Paul Tyler, Faezzah Baharom, Tao Sun, Eva Lin, Scott Martin, Brandon D Kayser, Robert J Johnston, Ira Mellman, Lélia Delamarre, Nathaniel R West, Sören Müller, Yan Qu, Klaus Heger

The DNA exonuclease three-prime repair exonuclease 1 (TREX1) is critical for preventing autoimmunity in mice and humans by degrading endogenous cytosolic DNA, which otherwise triggers activation of the innate cGAS/STING pathway leading to the production of type I IFNs. As tumor cells are prone to aberrant cytosolic DNA accumulation, we hypothesized that they are critically dependent on TREX1 activity to limit their immunogenicity. Here, we show that in tumor cells, TREX1 restricts spontaneous activation of the cGAS/STING pathway, and the subsequent induction of a type I IFN response. As a result, TREX1 deficiency compromised in vivo tumor growth in mice. This delay in tumor growth depended on a functional immune system, systemic type I IFN signaling, and tumor-intrinsic cGAS expression. Mechanistically, we show that tumor TREX1 loss drove activation of CD8+ T cells and NK cells, prevented CD8+ T-cell exhaustion, and remodeled an immunosuppressive myeloid compartment. Consequently, TREX1 deficiency combined with T-cell-directed immune checkpoint blockade. Collectively, we conclude that TREX1 is essential to limit tumor immunogenicity, and that targeting this innate immune checkpoint remodels the tumor microenvironment and enhances antitumor immunity by itself and in combination with T-cell-targeted therapies. See related article by Toufektchan et al., p. 673.

DNA外切酶TREX1(Three-prime repair exonuclease 1,三价修复外切酶1)通过降解内源性细胞膜DNA来防止小鼠和人类的自身免疫,否则会引发先天性cGAS/STING途径的激活,导致I型IFNs的产生。由于肿瘤细胞容易出现异常的细胞膜 DNA 积累,我们推测它们严重依赖 TREX1 的活性来限制其免疫原性。我们在这里发现,在肿瘤细胞中,TREX1 确实限制了 cGAS/STING 通路的自发激活以及随后 I 型 IFN 反应的诱导。因此,TREX1的缺乏会影响小鼠体内肿瘤的生长。这种延迟取决于功能性免疫系统、系统性 I 型 IFN 信号传导和肿瘤内在 cGAS 表达。从机理上讲,我们发现肿瘤 TREX1 的缺失会促进 CD8 T 细胞和 NK 细胞的活化,防止 CD8 T 细胞衰竭,并重塑免疫抑制性髓细胞区系。因此,TREX1的缺失与T细胞导向的免疫检查点阻断具有协同作用。综上所述,我们得出结论:TREX1 对限制肿瘤免疫原性至关重要,靶向这一先天性免疫检查点可重塑肿瘤微环境,并通过自身或与 T 细胞靶向疗法相结合增强抗肿瘤免疫力。
{"title":"The Exonuclease TREX1 Constitutes an Innate Immune Checkpoint Limiting cGAS/STING-Mediated Antitumor Immunity.","authors":"Junghyun Lim, Ryan Rodriguez, Katherine Williams, John Silva, Alan G Gutierrez, Paul Tyler, Faezzah Baharom, Tao Sun, Eva Lin, Scott Martin, Brandon D Kayser, Robert J Johnston, Ira Mellman, Lélia Delamarre, Nathaniel R West, Sören Müller, Yan Qu, Klaus Heger","doi":"10.1158/2326-6066.CIR-23-1078","DOIUrl":"10.1158/2326-6066.CIR-23-1078","url":null,"abstract":"<p><p>The DNA exonuclease three-prime repair exonuclease 1 (TREX1) is critical for preventing autoimmunity in mice and humans by degrading endogenous cytosolic DNA, which otherwise triggers activation of the innate cGAS/STING pathway leading to the production of type I IFNs. As tumor cells are prone to aberrant cytosolic DNA accumulation, we hypothesized that they are critically dependent on TREX1 activity to limit their immunogenicity. Here, we show that in tumor cells, TREX1 restricts spontaneous activation of the cGAS/STING pathway, and the subsequent induction of a type I IFN response. As a result, TREX1 deficiency compromised in vivo tumor growth in mice. This delay in tumor growth depended on a functional immune system, systemic type I IFN signaling, and tumor-intrinsic cGAS expression. Mechanistically, we show that tumor TREX1 loss drove activation of CD8+ T cells and NK cells, prevented CD8+ T-cell exhaustion, and remodeled an immunosuppressive myeloid compartment. Consequently, TREX1 deficiency combined with T-cell-directed immune checkpoint blockade. Collectively, we conclude that TREX1 is essential to limit tumor immunogenicity, and that targeting this innate immune checkpoint remodels the tumor microenvironment and enhances antitumor immunity by itself and in combination with T-cell-targeted therapies. See related article by Toufektchan et al., p. 673.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"663-672"},"PeriodicalIF":8.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11148535/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140136498","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The ω-3 Polyunsaturated Fatty Acid Docosahexaenoic Acid Enhances NK-Cell Antitumor Effector Functions. ω-3多不饱和脂肪酸二十二碳六烯酸能增强NK细胞的抗肿瘤效应功能。
IF 10.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-23-0359
Shuting Wu, Hongyan Peng, Songyang Li, Lanlan Huang, Xiangyu Wang, Yana Li, Yongjie Liu, Peiwen Xiong, Qinglan Yang, Kunpeng Tian, Weiru Wu, Rongxi Pu, Xiulan Lu, Zhenghui Xiao, Jian Yang, Zhaoyang Zhong, Yuan Gao, Yafei Deng, Youcai Deng

ω-3 polyunsaturated fatty acids (PUFA) are known to directly repress tumor development and progression. In this study, we explored whether docosahexaenoic acid (DHA), a type of ω-3 PUFA, had an immunomodulatory role in inhibiting tumor growth in immunocompetent mice. The number of natural killer (NK) cells but not the number of T or B cells was decreased by DHA supplementation in various tissues under physiologic conditions. Although the frequency and number of NK cells were comparable, IFNγ production by NK cells in both the spleen and lung was increased in DHA-supplemented mice in the mouse B16F10 melanoma tumor model. Single-cell RNA sequencing revealed that DHA promoted effector function and oxidative phosphorylation in NK cells but had no obvious effects on other immune cells. Using Rag2-/- mice and NK-cell depletion by PK136 antibody injection, we demonstrated that the suppression of B16F10 melanoma tumor growth in the lung by DHA supplementation was dependent mainly on NK cells. In vitro experiments showed that DHA directly enhanced IFNγ production, CD107a expression, and mitochondrial oxidative phosphorylation (OXPHOS) activity and slightly increased proliferator-activated receptor gamma coactivator-1α (PGC-1α) protein expression in NK cells. The PGC-1α inhibitor SR-18292 in vitro and NK cell-specific knockout of PGC-1α in mice reversed the antitumor effects of DHA. In summary, our findings broaden the current knowledge on how DHA supplementation protects against cancer growth from the perspective of immunomodulation by upregulating PGC-1α signaling-mediated mitochondrial OXPHOS activity in NK cells.

众所周知,ω-3 多不饱和脂肪酸(PUFA)可直接抑制肿瘤的发展和恶化。在这项研究中,我们探讨了二十二碳六烯酸(DHA)(一种ω-3多不饱和脂肪酸)在促进免疫功能正常的小鼠肿瘤生长方面是否具有免疫调节作用。在生理条件下,各种组织中补充 DHA 会减少自然杀伤细胞(NK)的数量,但不会减少 T 细胞或 B 细胞的数量。在小鼠 B16F10 黑色素瘤模型中,虽然 NK 细胞的频率和数量相当,但补充 DHA 的小鼠脾脏和肺部的 NK 细胞产生的 IFN-γ 均有所增加。单细胞 RNA 测序(scRNA-seq)显示,DHA 促进了 NK 细胞的效应功能和氧化磷酸化,但对其他免疫细胞没有明显影响。利用Rag2-/-小鼠和通过注射PK136抗体消耗NK细胞,我们证明了补充DHA对肺部B16F10黑色素瘤生长的抑制主要依赖于NK细胞。体外实验表明,DHA能直接增强NK细胞中IFN-γ的产生、CD107a的表达和线粒体氧化磷酸化(OXPHOS)活性,并能轻微增加增殖体激活受体γ辅激活剂-1α(PGC-1α)蛋白的表达。体外 PGC-1α 抑制剂 SR-18292 和小鼠 NK 细胞特异性 PGC-1α 基因敲除可逆转 DHA 的抗肿瘤作用。总之,我们的研究结果从免疫调节的角度拓宽了目前关于补充 DHA 如何通过上调 NK 细胞中 PGC-1α 信号介导的线粒体 OXPHOS 活性来保护癌症生长的知识。
{"title":"The ω-3 Polyunsaturated Fatty Acid Docosahexaenoic Acid Enhances NK-Cell Antitumor Effector Functions.","authors":"Shuting Wu, Hongyan Peng, Songyang Li, Lanlan Huang, Xiangyu Wang, Yana Li, Yongjie Liu, Peiwen Xiong, Qinglan Yang, Kunpeng Tian, Weiru Wu, Rongxi Pu, Xiulan Lu, Zhenghui Xiao, Jian Yang, Zhaoyang Zhong, Yuan Gao, Yafei Deng, Youcai Deng","doi":"10.1158/2326-6066.CIR-23-0359","DOIUrl":"10.1158/2326-6066.CIR-23-0359","url":null,"abstract":"<p><p>ω-3 polyunsaturated fatty acids (PUFA) are known to directly repress tumor development and progression. In this study, we explored whether docosahexaenoic acid (DHA), a type of ω-3 PUFA, had an immunomodulatory role in inhibiting tumor growth in immunocompetent mice. The number of natural killer (NK) cells but not the number of T or B cells was decreased by DHA supplementation in various tissues under physiologic conditions. Although the frequency and number of NK cells were comparable, IFNγ production by NK cells in both the spleen and lung was increased in DHA-supplemented mice in the mouse B16F10 melanoma tumor model. Single-cell RNA sequencing revealed that DHA promoted effector function and oxidative phosphorylation in NK cells but had no obvious effects on other immune cells. Using Rag2-/- mice and NK-cell depletion by PK136 antibody injection, we demonstrated that the suppression of B16F10 melanoma tumor growth in the lung by DHA supplementation was dependent mainly on NK cells. In vitro experiments showed that DHA directly enhanced IFNγ production, CD107a expression, and mitochondrial oxidative phosphorylation (OXPHOS) activity and slightly increased proliferator-activated receptor gamma coactivator-1α (PGC-1α) protein expression in NK cells. The PGC-1α inhibitor SR-18292 in vitro and NK cell-specific knockout of PGC-1α in mice reversed the antitumor effects of DHA. In summary, our findings broaden the current knowledge on how DHA supplementation protects against cancer growth from the perspective of immunomodulation by upregulating PGC-1α signaling-mediated mitochondrial OXPHOS activity in NK cells.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"744-758"},"PeriodicalIF":10.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11148550/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140206337","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer immunology research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1