首页 > 最新文献

Cancer immunology research最新文献

英文 中文
RNA-encoded Interleukin 2 with Extended Bioavailability Amplifies RNA Vaccine-Induced Antitumor T-cell Immunity. 具有扩展生物利用度的 RNA 编码白细胞介素-2 可增强 RNA 疫苗诱导的抗肿瘤 T 细胞免疫力。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-01 DOI: 10.1158/2326-6066.CIR-23-0701
Daniel Peters, Lena M Kranz, David Eisel, Mustafa Diken, Sebastian Kreiter, Özlem Türeci, Ugur Sahin, Mathias Vormehr

Interleukin 2 (IL-2) is a crucial cytokine in T-cell immunity, with a promising potential in cancer vaccines. However, therapeutic application of IL-2 is hampered by its short half-life and substantial toxicity. This study reports preclinical characterization of a mouse serum albumin-IL-2 fusion protein (Alb-IL2) encoded on nucleoside-modified RNA that is delivered via a nanoparticle formulation (Alb-IL2 RNA-NP) mediating prolonged cytokine availability. Alb-IL2 RNA-NP was combined with RNA-lipoplex (RNA-LPX) vaccines to evaluate its effect on the expansion of vaccine-induced antigen specific T-cell immunity. In mice dosed with Alb-IL2 RNA-NP, translated protein was shown to be systemically available up to 2 days, with an albumin-dependent preferred presence in the tumor and tumor-draining lymph node. Alb-IL2 RNA-NP administration prolonged serum availability of the cytokine compared with murine recombinant IL-2. In combination with RNA-LPX vaccines, Alb-IL2 RNA-NP administration highly increased the expansion of RNA-LPX vaccine-induced CD8+ T cells in the spleen and blood. The combination enhanced and sustained the fraction of IL-2 receptor (IL-2R) α-positive antigen-specific CD8+ T cells and ameliorated the functional capacity of the CD8+ T-cell population. Alb-IL2 RNA-NP strongly improved the antitumor activity and survival of concomitant RNA-LPX vaccination and PD-L1 blockade in a subcutaneous mouse tumor model. The favorable pharmacokinetic properties of Alb-IL2 RNA-NP render it an attractive modality for rationally designed combination immunotherapy. RNA vaccines that induce tumor-specific T-cell immunity for Alb-IL2 RNA-NP to further amplify are particularly attractive combination partners.

白细胞介素-2(IL-2)是 T 细胞免疫中的一种重要细胞因子,也是一种很有希望提高癌症疫苗疗效的组合伙伴。然而,IL-2 的半衰期短、毒性大,阻碍了它的治疗应用。在此,我们报告了一种小鼠血清白蛋白-IL-2融合蛋白(Alb-IL2)的临床前特征,该蛋白由核苷修饰的RNA编码,通过纳米颗粒制剂(Alb-IL2 RNA-NP)递送,可延长细胞因子的可用性。Alb-IL2 RNA-NP 与 RNA-脂质体(RNA-LPX)疫苗结合使用,以评估其对扩大疫苗诱导的抗原特异性 T 细胞免疫的影响。研究表明,在服用 Alb-IL2 RNA-NP 的小鼠体内,翻译蛋白可在两天内全身存在,肿瘤和肿瘤引流淋巴结中的白蛋白依赖性较强。与小鼠重组IL-2(rIL-2)相比,Alb-IL2 RNA-NP可延长细胞因子在血清中的存在时间。Alb-IL2 RNA-NP与RNA-LPX疫苗联合使用,可显著提高RNA-LPX疫苗诱导的CD8+T细胞在脾脏和血液中的扩增。联合用药可提高并维持 IL-2 受体(IL-2R)α 阳性的抗原特异性 CD8+ T 细胞的比例,并改善 CD8+ T 细胞群的功能能力。在小鼠皮下肿瘤模型中,Alb-IL2 RNA-NP能显著提高同时接种RNA-LPX疫苗和PD-L1阻断剂的抗肿瘤活性和存活率。Alb-IL2 RNA-NP 良好的药代动力学特性使其成为合理设计联合免疫疗法的一种有吸引力的模式。能诱导肿瘤特异性 T 细胞免疫,使 Alb-IL2 RNA-NP 进一步放大的 RNA 疫苗是特别有吸引力的组合伙伴。
{"title":"RNA-encoded Interleukin 2 with Extended Bioavailability Amplifies RNA Vaccine-Induced Antitumor T-cell Immunity.","authors":"Daniel Peters, Lena M Kranz, David Eisel, Mustafa Diken, Sebastian Kreiter, Özlem Türeci, Ugur Sahin, Mathias Vormehr","doi":"10.1158/2326-6066.CIR-23-0701","DOIUrl":"10.1158/2326-6066.CIR-23-0701","url":null,"abstract":"<p><p>Interleukin 2 (IL-2) is a crucial cytokine in T-cell immunity, with a promising potential in cancer vaccines. However, therapeutic application of IL-2 is hampered by its short half-life and substantial toxicity. This study reports preclinical characterization of a mouse serum albumin-IL-2 fusion protein (Alb-IL2) encoded on nucleoside-modified RNA that is delivered via a nanoparticle formulation (Alb-IL2 RNA-NP) mediating prolonged cytokine availability. Alb-IL2 RNA-NP was combined with RNA-lipoplex (RNA-LPX) vaccines to evaluate its effect on the expansion of vaccine-induced antigen specific T-cell immunity. In mice dosed with Alb-IL2 RNA-NP, translated protein was shown to be systemically available up to 2 days, with an albumin-dependent preferred presence in the tumor and tumor-draining lymph node. Alb-IL2 RNA-NP administration prolonged serum availability of the cytokine compared with murine recombinant IL-2. In combination with RNA-LPX vaccines, Alb-IL2 RNA-NP administration highly increased the expansion of RNA-LPX vaccine-induced CD8+ T cells in the spleen and blood. The combination enhanced and sustained the fraction of IL-2 receptor (IL-2R) α-positive antigen-specific CD8+ T cells and ameliorated the functional capacity of the CD8+ T-cell population. Alb-IL2 RNA-NP strongly improved the antitumor activity and survival of concomitant RNA-LPX vaccination and PD-L1 blockade in a subcutaneous mouse tumor model. The favorable pharmacokinetic properties of Alb-IL2 RNA-NP render it an attractive modality for rationally designed combination immunotherapy. RNA vaccines that induce tumor-specific T-cell immunity for Alb-IL2 RNA-NP to further amplify are particularly attractive combination partners.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1409-1420"},"PeriodicalIF":8.1,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141417946","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
Targeting Tumor-Associated Sialic Acids Using Chimeric Switch Receptors Based on Siglec-9 Enhances the Antitumor Efficacy of Engineered T Cells. 利用基于 Siglec-9 的嵌合开关受体靶向肿瘤相关的 Sialic 酸可增强工程 T 细胞的抗肿瘤功效
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-01 DOI: 10.1158/2326-6066.CIR-23-0823
Vasyl Eisenberg, Shiran Hoogi, Erel Katzman, Nimrod Ben Haim, Raphaelle Zur-Toledano, Maria Radman, Yishai Reboh, Oranit Zadok, Iris Kamer, Jair Bar, Irit Sagi, Ayal Hendel, Cyrille J Cohen

Cancer exploits different mechanisms to escape T-cell immunosurveillance, including overexpression of checkpoint ligands, secretion of immunosuppressive molecules, and aberrant glycosylation. Herein, we report that IFNγ, a potent immunomodulator secreted in the tumor microenvironment, can induce α2,6 hypersialylation in cancer cell lines derived from various histologies. We focused on Siglec-9, a receptor for sialic acid moieties, and demonstrated that the Siglec-9+ T-cell population displayed reduced effector function. We speculated that Siglec-9 in primary human T cells can act as a checkpoint molecule and demonstrated that knocking out Siglec-9 using a clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system enhanced the functionality of primary human T cells. Finally, we aimed to augment cancer-specific T-cell activity by taking advantage of tumor hypersialylation. Thus, we designed several Siglec-9-based chimeric switch receptors (CSR), which included an intracellular moiety derived from costimulatory molecules (CD28/41BB) and different hinge regions. In an antigen-specific context, T cells transduced with Siglec-9 CSRs demonstrated increased cytokine secretions and upregulation of activation markers. Moreover, T cells equipped with specific Siglec-9 CSRs mediated robust antitumor activity in a xenograft model of human tumors. Overall, this work sheds light on tumor evasion mechanisms mediated by sialylated residues and exemplifies an approach to improve engineered T cell-based cancer treatment. See related Spotlight by Abken, p. 1310.

癌症利用不同的机制逃避 T 细胞的免疫监视,包括检查点配体的过度表达、免疫抑制分子的分泌和异常糖基化。在本文中,我们报告了在肿瘤微环境中分泌的强效免疫调节剂 IFNγ 可诱导不同组织结构的癌细胞株中α2,6 过度糖基化。我们随后重点研究了Siglec-9--一种针对sialic acid分子的受体,结果表明Siglec-9+ T细胞群的效应功能降低了。我们推测原代人类 T 细胞中的 Siglec-9 可充当检查点分子,并证明使用 CRISPR/Cas9 系统敲除 Siglec-9 可增强原代人类 T 细胞的功能。最后,我们的目标是利用肿瘤的高ialylation 增强癌症特异性 T 细胞的活性。因此,我们设计了几种基于 Siglec-9 的嵌合开关受体 (CSR),其中包括源自成本刺激分子(CD28/41BB)的细胞内分子和不同的铰链区。在抗原特异性的情况下,用 Siglec-9 CSRs 转导的 T 细胞表现出细胞因子分泌增加和活化标志物上调。此外,在人类肿瘤的异种移植模型中,装有特异性 Siglec-9 CSRs 的 T 细胞具有很强的抗肿瘤活性。总之,这项工作揭示了由硅戊基化残基介导的肿瘤逃避机制,并为改进基于工程 T 细胞的癌症治疗提供了一种方法。
{"title":"Targeting Tumor-Associated Sialic Acids Using Chimeric Switch Receptors Based on Siglec-9 Enhances the Antitumor Efficacy of Engineered T Cells.","authors":"Vasyl Eisenberg, Shiran Hoogi, Erel Katzman, Nimrod Ben Haim, Raphaelle Zur-Toledano, Maria Radman, Yishai Reboh, Oranit Zadok, Iris Kamer, Jair Bar, Irit Sagi, Ayal Hendel, Cyrille J Cohen","doi":"10.1158/2326-6066.CIR-23-0823","DOIUrl":"10.1158/2326-6066.CIR-23-0823","url":null,"abstract":"<p><p>Cancer exploits different mechanisms to escape T-cell immunosurveillance, including overexpression of checkpoint ligands, secretion of immunosuppressive molecules, and aberrant glycosylation. Herein, we report that IFNγ, a potent immunomodulator secreted in the tumor microenvironment, can induce α2,6 hypersialylation in cancer cell lines derived from various histologies. We focused on Siglec-9, a receptor for sialic acid moieties, and demonstrated that the Siglec-9+ T-cell population displayed reduced effector function. We speculated that Siglec-9 in primary human T cells can act as a checkpoint molecule and demonstrated that knocking out Siglec-9 using a clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system enhanced the functionality of primary human T cells. Finally, we aimed to augment cancer-specific T-cell activity by taking advantage of tumor hypersialylation. Thus, we designed several Siglec-9-based chimeric switch receptors (CSR), which included an intracellular moiety derived from costimulatory molecules (CD28/41BB) and different hinge regions. In an antigen-specific context, T cells transduced with Siglec-9 CSRs demonstrated increased cytokine secretions and upregulation of activation markers. Moreover, T cells equipped with specific Siglec-9 CSRs mediated robust antitumor activity in a xenograft model of human tumors. Overall, this work sheds light on tumor evasion mechanisms mediated by sialylated residues and exemplifies an approach to improve engineered T cell-based cancer treatment. See related Spotlight by Abken, p. 1310.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1380-1391"},"PeriodicalIF":8.1,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141733568","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
Transforming the Dark into Light: A Siglec-9 Switch. 化暗为明:Siglec-9 开关。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-01 DOI: 10.1158/2326-6066.CIR-24-0429
Hinrich Abken

Tumor-associated immune repression dampens the success of T-cell therapy for cancer by a plethora of inhibitory mechanisms including aberrant glycosylation. In this issue, Eisenberg and colleagues show that IFNγ induces hyper-sialylation of cancer cells and that this acts as the "checkpoint" through binding to the inhibitory molecule Siglec-9 on immune cells. A chimeric Siglec-9 "switch" receptor converts the suppressive signal into a stimulatory signal, thereby restoring T-cell responses in the tumor tissue, which has multiple implications for the use of adoptive cell therapy in cancer. See related article by Eisenberg et al., p. 1380 (3).

肿瘤相关的免疫抑制通过包括异常糖基化在内的多种抑制机制抑制了T细胞疗法治疗癌症的成功。在本期杂志中,艾森伯格及其同事发现,IFNγ会诱导癌细胞的过度糖基化,并通过与免疫细胞上的抑制分子Siglec-9结合,起到 "检查点 "的作用。一种嵌合的 Siglec-9 "开关 "受体能将抑制信号转换为刺激信号,从而恢复肿瘤组织中的 T 细胞反应,这对癌症的采用性细胞疗法具有多方面的意义。见 Eisenberg 等人的相关文章,第 XX (3) 页。
{"title":"Transforming the Dark into Light: A Siglec-9 Switch.","authors":"Hinrich Abken","doi":"10.1158/2326-6066.CIR-24-0429","DOIUrl":"10.1158/2326-6066.CIR-24-0429","url":null,"abstract":"<p><p>Tumor-associated immune repression dampens the success of T-cell therapy for cancer by a plethora of inhibitory mechanisms including aberrant glycosylation. In this issue, Eisenberg and colleagues show that IFNγ induces hyper-sialylation of cancer cells and that this acts as the \"checkpoint\" through binding to the inhibitory molecule Siglec-9 on immune cells. A chimeric Siglec-9 \"switch\" receptor converts the suppressive signal into a stimulatory signal, thereby restoring T-cell responses in the tumor tissue, which has multiple implications for the use of adoptive cell therapy in cancer. See related article by Eisenberg et al., p. 1380 (3).</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1310"},"PeriodicalIF":8.1,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141733569","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 : 2024-10-01 DOI: 10.1158/2326-6066.CIR-12-10-WWR
{"title":"A Sampling of Highlights from the Literature.","authors":"","doi":"10.1158/2326-6066.CIR-12-10-WWR","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-12-10-WWR","url":null,"abstract":"","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":"12 10","pages":"1309"},"PeriodicalIF":8.1,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142342195","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 E3 Ubiquitin Ligase FBXO38 Maintains the Antitumor Function of Natural Killer Cells by Sustaining IL15R Signaling. E3泛素连接酶FBXO38通过维持IL-15R信号维持自然杀伤细胞的抗肿瘤功能。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-01 DOI: 10.1158/2326-6066.CIR-23-1061
Yongjing Shi, Xiaodong Zheng, Hui Peng, Chenqi Xu, Rui Sun, Zhigang Tian, Haoyu Sun, Xianwei Wang

Natural killer (NK) cells are the main innate antitumor effector cells but their function is often constrained in the tumor microenvironment. It has been reported that the E3 ligase FBXO38 accelerates PD-1 degradation in tumor-infiltrating T cells to unleash their cytotoxic function. In this study, we found that the transcriptional levels of FBXO38 in intratumoral NK cells of patients with cancer and tumor-bearing mice were significantly lower than in peritumoral NK cells. Conditional knockout of FBXO38 in NK cells accelerated tumor growth and increased tumor metastasis. FBXO38 deficiency resulted in impaired proliferation and survival of tumor-infiltrating NK (TINK) cells. Mechanistically, FBXO38 deficiency enhanced TGF-β signaling, including elevating expression of Smad2 and Smad3, which suppressed expression of the transcription factor Eomes and further reduced expression of surface IL15Rβ and IL15Rγc on NK cells. Consequently, FBXO38 deficiency led to TINK cell hyporesponsiveness to IL15. Consistent with these observations, FBXO38 mRNA expression was positively correlated with the proliferation of TINK cells in multiple human tumors. To study the therapeutic potential of FBXO38, mice bearing human tumors were treated with FBXO38 overexpressed human primary NK cells and showed a significant reduction in tumor size and prolonged survival. In conclusion, our results suggest that FBXO38 sustains NK-cell expansion and survival to promote antitumor immunity and have potential therapeutic implications as they suggest FBXO38 could be harnessed to enhance NK cell-based cancer immunotherapy.

自然杀伤(NK)细胞是主要的先天性抗肿瘤效应细胞,但其功能在肿瘤微环境(TME)中往往受到限制。有报道称,E3连接酶FBXO38能加速肿瘤浸润T细胞中PD-1的降解,从而释放其细胞毒性功能。在这项研究中,我们发现癌症患者和肿瘤小鼠瘤内 NK 细胞中的 FBXO38 转录水平明显低于瘤周 NK 细胞。条件性敲除(cKO)NK细胞中的FBXO38会加速肿瘤生长并增加肿瘤转移。缺乏 FBXO38 会导致肿瘤浸润 NK(TINK)细胞的增殖和存活能力受损。从机制上讲,FBXO38 缺乏会增强 TGF-β 信号传导,包括提高 Smad2 和 Smad3 的表达,从而抑制转录因子 Eomes 的表达,进一步降低 NK 细胞表面 IL-15Rβ 和 IL-15Rγc 的表达。因此,FBXO38 的缺乏会导致 TINK 细胞对 IL-15 的低反应性。与这些观察结果一致的是,在多种人类肿瘤中,FBXO38 mRNA 的表达与 TINK 细胞的增殖呈正相关。为了研究 FBXO38 的治疗潜力,用过表达 FBXO38 的人类原代 NK 细胞治疗携带人类肿瘤的小鼠,结果显示肿瘤体积显著缩小,存活时间延长。总之,我们的研究结果表明,FBXO38 可维持 NK 细胞的扩增和存活,从而促进抗肿瘤免疫,并具有潜在的治疗意义,因为它们表明可以利用 FBXO38 来增强基于 NK 细胞的癌症免疫疗法。
{"title":"The E3 Ubiquitin Ligase FBXO38 Maintains the Antitumor Function of Natural Killer Cells by Sustaining IL15R Signaling.","authors":"Yongjing Shi, Xiaodong Zheng, Hui Peng, Chenqi Xu, Rui Sun, Zhigang Tian, Haoyu Sun, Xianwei Wang","doi":"10.1158/2326-6066.CIR-23-1061","DOIUrl":"10.1158/2326-6066.CIR-23-1061","url":null,"abstract":"<p><p>Natural killer (NK) cells are the main innate antitumor effector cells but their function is often constrained in the tumor microenvironment. It has been reported that the E3 ligase FBXO38 accelerates PD-1 degradation in tumor-infiltrating T cells to unleash their cytotoxic function. In this study, we found that the transcriptional levels of FBXO38 in intratumoral NK cells of patients with cancer and tumor-bearing mice were significantly lower than in peritumoral NK cells. Conditional knockout of FBXO38 in NK cells accelerated tumor growth and increased tumor metastasis. FBXO38 deficiency resulted in impaired proliferation and survival of tumor-infiltrating NK (TINK) cells. Mechanistically, FBXO38 deficiency enhanced TGF-β signaling, including elevating expression of Smad2 and Smad3, which suppressed expression of the transcription factor Eomes and further reduced expression of surface IL15Rβ and IL15Rγc on NK cells. Consequently, FBXO38 deficiency led to TINK cell hyporesponsiveness to IL15. Consistent with these observations, FBXO38 mRNA expression was positively correlated with the proliferation of TINK cells in multiple human tumors. To study the therapeutic potential of FBXO38, mice bearing human tumors were treated with FBXO38 overexpressed human primary NK cells and showed a significant reduction in tumor size and prolonged survival. In conclusion, our results suggest that FBXO38 sustains NK-cell expansion and survival to promote antitumor immunity and have potential therapeutic implications as they suggest FBXO38 could be harnessed to enhance NK cell-based cancer immunotherapy.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1438-1451"},"PeriodicalIF":8.1,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141579027","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
Hypoxia-Induced Long Noncoding RNA HIF1A-AS2 Regulates Stability of MHC Class I Protein in Head and Neck Cancer. 缺氧诱导的长非编码 RNA HIF1A-AS2 调节头颈癌中 MHC I 类蛋白的稳定性
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-01 DOI: 10.1158/2326-6066.CIR-23-0622
Tsai-Tsen Liao, Yu-Hsien Chen, Zih-Yu Li, An-Ching Hsiao, Ya-Li Huang, Ruo-Xin Hao, Shyh-Kuan Tai, Pen-Yuan Chu, Jing-Wen Shih, Hsing-Jien Kung, Muh-Hwa Yang

Intratumoral hypoxia not only promotes angiogenesis and invasiveness of cancer cells but also creates an immunosuppressive microenvironment that facilitates tumor progression. However, the mechanisms by which hypoxic tumor cells disseminate immunosuppressive signals remain unclear. In this study, we demonstrate that a hypoxia-induced long noncoding RNA HIF1A Antisense RNA 2 (HIF1A-AS2) is upregulated in hypoxic tumor cells and hypoxic tumor-derived exosomes in head and neck squamous cell carcinoma (HNSCC). Hypoxia-inducible factor 1 alpha (HIF1α) was found to directly bind to the regulatory region of HIF1A-AS2 to enhance its expression. HIF1A-AS2 reduced the protein stability of major histocompatibility complex class I (MHC-I) by promoting the interaction between the autophagy cargo receptor neighbor of BRCA1 gene 1 (NBR1) protein and MHC-I, thereby increasing the autophagic degradation of MHC-I. In HNSCC samples, the expression of HIF1A-AS2 was found to correlate with hypoxic signatures and advanced clinical stages. Patients with high HIF1α and low HLA-ABC expression showed reduced infiltration of CD8+ T cells. These findings define a mechanism of hypoxia-mediated immune evasion in HNSCC through downregulation of antigen-presenting machinery via intracellular or externalized hypoxia-induced long noncoding RNA.

瘤内缺氧不仅会促进血管生成和癌细胞的侵袭性,还会形成一种免疫抑制微环境,从而促进肿瘤的进展。然而,低氧肿瘤细胞传播免疫抑制信号的机制仍不清楚。在这项研究中,我们发现在头颈部鳞状细胞癌(HNSCC)中,低氧诱导的长非编码 RNA(lncRNA)HIF1A 反义 RNA 2(HIF1A-AS2)在低氧肿瘤细胞和低氧肿瘤衍生外泌体(TEXs)中均上调。研究发现,低氧诱导因子1α1(HIF-1α)可直接与HIF1A-AS2的调节区结合,从而增强其表达。HIF1A-AS2通过促进自噬货物受体Neighbor of BRCA1 gene 1蛋白(NBR1)与MHC-I之间的相互作用,从而增加MHC-I的自噬降解,降低了主要组织相容性复合体I类(MHC-I)蛋白的稳定性。在 HNSCC 样本中,HIF1A-AS2 的表达与缺氧特征和晚期临床分期相关。高HIF-1α和低HLA-ABC表达的患者显示CD8+ T细胞浸润减少。这些发现确定了HNSCC中缺氧介导的免疫逃避机制,即通过细胞内或外化的缺氧诱导lncRNA下调抗原递呈机制。
{"title":"Hypoxia-Induced Long Noncoding RNA HIF1A-AS2 Regulates Stability of MHC Class I Protein in Head and Neck Cancer.","authors":"Tsai-Tsen Liao, Yu-Hsien Chen, Zih-Yu Li, An-Ching Hsiao, Ya-Li Huang, Ruo-Xin Hao, Shyh-Kuan Tai, Pen-Yuan Chu, Jing-Wen Shih, Hsing-Jien Kung, Muh-Hwa Yang","doi":"10.1158/2326-6066.CIR-23-0622","DOIUrl":"10.1158/2326-6066.CIR-23-0622","url":null,"abstract":"<p><p>Intratumoral hypoxia not only promotes angiogenesis and invasiveness of cancer cells but also creates an immunosuppressive microenvironment that facilitates tumor progression. However, the mechanisms by which hypoxic tumor cells disseminate immunosuppressive signals remain unclear. In this study, we demonstrate that a hypoxia-induced long noncoding RNA HIF1A Antisense RNA 2 (HIF1A-AS2) is upregulated in hypoxic tumor cells and hypoxic tumor-derived exosomes in head and neck squamous cell carcinoma (HNSCC). Hypoxia-inducible factor 1 alpha (HIF1α) was found to directly bind to the regulatory region of HIF1A-AS2 to enhance its expression. HIF1A-AS2 reduced the protein stability of major histocompatibility complex class I (MHC-I) by promoting the interaction between the autophagy cargo receptor neighbor of BRCA1 gene 1 (NBR1) protein and MHC-I, thereby increasing the autophagic degradation of MHC-I. In HNSCC samples, the expression of HIF1A-AS2 was found to correlate with hypoxic signatures and advanced clinical stages. Patients with high HIF1α and low HLA-ABC expression showed reduced infiltration of CD8+ T cells. These findings define a mechanism of hypoxia-mediated immune evasion in HNSCC through downregulation of antigen-presenting machinery via intracellular or externalized hypoxia-induced long noncoding RNA.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1468-1484"},"PeriodicalIF":8.1,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11443317/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141449756","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
A Phase 1 Trial of Trebananib, an Angiopoietin 1 and 2 Neutralizing Peptibody, Combined with Pembrolizumab in Patients with Advanced Ovarian and Colorectal Cancer. 血管生成素 1 和 2 中和肽抗体 Trebananib 与 Pembrolizumab 联合治疗晚期卵巢癌和结直肠癌患者的 1 期试验。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-30 DOI: 10.1158/2326-6066.CIR-23-1027
Brandon M Huffman, Osama E Rahma, Kevin Tyan, Yvonne Y Li, Anita Giobbie-Hurder, Benjamin L Schlechter, Bruno Bockorny, Michael P Manos, Andrew D Cherniack, Joanna Baginska, Adrian Mariño-Enríquez, Katrina Z Kao, Anna K Maloney, Allison Ferro, Sarah Kelland, Kimmie Ng, Harshabad Singh, Emma L Welsh, Kathleen L Pfaff, Marios Giannakis, Scott J Rodig, F Stephen Hodi, James M Cleary

Ovarian cancers and microsatellite stable (MSS) colorectal cancers (CRC) are insensitive to anti-PD1 immunotherapy, and new immunotherapeutic approaches are needed. Preclinical data suggests a relationship between immunotherapy resistance and elevated angiopoietin 2 levels. We performed a phase 1 dose-escalation study of pembrolizumab and the angiopoietin 1/2 inhibitor trebananib (NCT03239145). This multicenter trial enrolled patients with metastatic ovarian cancer or MSS CRC. Trebananib was administered intravenously weekly for 12 weeks with 200 mg intravenous pembrolizumab every 3 weeks. The toxicity profile of this combination was manageable, and the protocol-defined highest dose level (trebananib 30 mg/kg weekly plus pembrolizumab 200 mg every 3 weeks) was declared the maximum tolerated dose. The objective response rate for all patients was 7.3% (90% confidence interval: 2.5-15.9%). Three patients with MSS CRC had durable responses for ≥3 years. One responding patient's CRC harbored a POLE mutation. The other two responding patients had left-sided CRCs with no baseline liver metastases, and genomic analysis revealed that they both had KRAS wild-type, ERBB2 amplified tumors. After development of acquired resistance, biopsy of one patient's KRAS wild-type, ERBB2 amplified tumor showed a substantial decline in tumor-associated T cells and an increase in immunosuppressive intratumoral macrophages. Future studies are needed to carefully assess whether clinicogenomic features, such as lack of liver metastases, ERBB2 amplification, and left-sided tumors, can predict increased sensitivity to PD1 immunotherapy combinations.

卵巢癌和微卫星稳定(MSS)结直肠癌对抗PD1免疫疗法不敏感,因此需要新的免疫治疗方法。临床前数据表明,免疫疗法耐药与血管生成素2水平升高之间存在关系。我们对pembrolizumab和血管生成素1/2抑制剂trebananib进行了1期剂量递增研究(NCT03239145)。这项多中心试验招募了转移性卵巢癌或 MSS CRC 患者。特瑞巴尼布每周静脉注射一次,持续 12 周,同时每 3 周静脉注射 200 毫克 pembrolizumab。这种联合用药的毒性是可控的,方案定义的最高剂量水平(曲巴尼布每周30毫克/千克加pembrolizumab每3周200毫克)被宣布为最大耐受剂量。所有患者的客观反应率为 7.3%(90% 置信区间:2.5-15.9%)。三名 MSS CRC 患者的持久应答时间≥3 年。其中一名应答患者的 CRC 存在 POLE 突变。另外两名应答患者的左侧 CRC 没有基线肝转移,基因组分析显示他们都患有 KRAS 野生型、ERBB2 扩增的肿瘤。出现获得性耐药后,一名患者的 KRAS 野生型、ERBB2 扩增肿瘤的活检结果显示,肿瘤相关的 T 细胞大幅减少,免疫抑制性瘤内巨噬细胞增加。未来的研究需要仔细评估临床基因组学特征(如无肝转移、ERBB2扩增和左侧肿瘤)是否能预测对PD1免疫疗法组合的敏感性增加。
{"title":"A Phase 1 Trial of Trebananib, an Angiopoietin 1 and 2 Neutralizing Peptibody, Combined with Pembrolizumab in Patients with Advanced Ovarian and Colorectal Cancer.","authors":"Brandon M Huffman, Osama E Rahma, Kevin Tyan, Yvonne Y Li, Anita Giobbie-Hurder, Benjamin L Schlechter, Bruno Bockorny, Michael P Manos, Andrew D Cherniack, Joanna Baginska, Adrian Mariño-Enríquez, Katrina Z Kao, Anna K Maloney, Allison Ferro, Sarah Kelland, Kimmie Ng, Harshabad Singh, Emma L Welsh, Kathleen L Pfaff, Marios Giannakis, Scott J Rodig, F Stephen Hodi, James M Cleary","doi":"10.1158/2326-6066.CIR-23-1027","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-23-1027","url":null,"abstract":"<p><p>Ovarian cancers and microsatellite stable (MSS) colorectal cancers (CRC) are insensitive to anti-PD1 immunotherapy, and new immunotherapeutic approaches are needed. Preclinical data suggests a relationship between immunotherapy resistance and elevated angiopoietin 2 levels. We performed a phase 1 dose-escalation study of pembrolizumab and the angiopoietin 1/2 inhibitor trebananib (NCT03239145). This multicenter trial enrolled patients with metastatic ovarian cancer or MSS CRC. Trebananib was administered intravenously weekly for 12 weeks with 200 mg intravenous pembrolizumab every 3 weeks. The toxicity profile of this combination was manageable, and the protocol-defined highest dose level (trebananib 30 mg/kg weekly plus pembrolizumab 200 mg every 3 weeks) was declared the maximum tolerated dose. The objective response rate for all patients was 7.3% (90% confidence interval: 2.5-15.9%). Three patients with MSS CRC had durable responses for ≥3 years. One responding patient's CRC harbored a POLE mutation. The other two responding patients had left-sided CRCs with no baseline liver metastases, and genomic analysis revealed that they both had KRAS wild-type, ERBB2 amplified tumors. After development of acquired resistance, biopsy of one patient's KRAS wild-type, ERBB2 amplified tumor showed a substantial decline in tumor-associated T cells and an increase in immunosuppressive intratumoral macrophages. Future studies are needed to carefully assess whether clinicogenomic features, such as lack of liver metastases, ERBB2 amplification, and left-sided tumors, can predict increased sensitivity to PD1 immunotherapy combinations.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.1,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142342192","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
Deleting Trim33 in myeloid cells improves the efficiency of radiotherapy through an interferon beta dependent anti-tumor immune response. 通过干扰素 beta 依赖性抗肿瘤免疫反应,删除骨髓细胞中的 Trim33 可提高放疗效率。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-26 DOI: 10.1158/2326-6066.CIR-24-0026
Anaïs Assouvie, Marine Gerbé De Thoré, Claire Torres, Véronique Ménard, Alexia Alfaro, Eric Deutsch, Michele Mondini, Germain Rousselet

Radiotherapy (RT) triggers an immune response that contributes to anti-tumor effects. Induction of interferon beta (IFN-β) is a key event in this immunogenicity of RT. We have previously shown that TRIM33, a chromatin reader, restrains IFN-β expression in Toll-like receptor-activated myeloid cells. Here, we explored whether deleting Trim33 in myeloid cells might improve the radio-induced immune response, and subsequent efficiency of RT. We first established that Trim33-/- bone marrow-derived macrophages showed increased expression of IFN-β in response to direct irradiation, or to treatment with irradiated cancer cells, further supporting our hypothesis. We then tested the efficiency of a single dose RT in three subcutaneous and one orthotopic tumor models. In all situations, myeloid deletion of Trim33 led to a significantly improved response after RT, leading to a complete and durable response in most of the treated mice bearing orthotopic oral tumors. This effect required the IFN-I pathway, and the presence of CD8+ T lymphocytes, but not NK cells. In addition, cured mice were capable of rejecting a secondary tumor challenge, demonstrating an in situ vaccination effect. We conclude that deleting Trim33 in myeloid cells improves RT efficiency, through a mechanism involving the IFN-I pathway and the immune response. Our work suggests that myeloid Trim33 is a host factor affecting the tumor response to RT, thus representing a new potential therapeutic target for modifying RT responses.

放疗(RT)会引发免疫反应,从而产生抗肿瘤效应。诱导干扰素 beta(IFN-β)是 RT 免疫原性的一个关键事件。我们之前已经证明,染色质阅读器 TRIM33 可抑制 IFN-β 在 Toll 样受体激活的骨髓细胞中的表达。在此,我们探讨了在髓系细胞中删除 Trim33 是否会改善放射诱导的免疫反应以及随后 RT 的效率。我们首先确定了 Trim33-/- 骨髓源性巨噬细胞在直接照射或照射癌细胞处理后 IFN-β 的表达增加,进一步支持了我们的假设。然后,我们在三个皮下肿瘤模型和一个正位肿瘤模型中测试了单剂量 RT 的效率。在所有情况下,Trim33的髓样体缺失都能显著改善RT后的反应,从而使大多数接受治疗的口腔正位肿瘤小鼠获得完全和持久的反应。这种效应需要 IFN-I 途径和 CD8+ T 淋巴细胞的存在,但不需要 NK 细胞。此外,治愈的小鼠能够拒绝二次肿瘤挑战,显示了原位疫苗接种效应。我们的结论是,通过涉及 IFN-I 通路和免疫反应的机制,删除髓系细胞中的 Trim33 可提高 RT 的效率。我们的研究表明,髓系细胞 Trim33 是影响肿瘤对 RT 反应的宿主因子,因此是改变 RT 反应的一个新的潜在治疗靶点。
{"title":"Deleting Trim33 in myeloid cells improves the efficiency of radiotherapy through an interferon beta dependent anti-tumor immune response.","authors":"Anaïs Assouvie, Marine Gerbé De Thoré, Claire Torres, Véronique Ménard, Alexia Alfaro, Eric Deutsch, Michele Mondini, Germain Rousselet","doi":"10.1158/2326-6066.CIR-24-0026","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-24-0026","url":null,"abstract":"<p><p>Radiotherapy (RT) triggers an immune response that contributes to anti-tumor effects. Induction of interferon beta (IFN-β) is a key event in this immunogenicity of RT. We have previously shown that TRIM33, a chromatin reader, restrains IFN-β expression in Toll-like receptor-activated myeloid cells. Here, we explored whether deleting Trim33 in myeloid cells might improve the radio-induced immune response, and subsequent efficiency of RT. We first established that Trim33-/- bone marrow-derived macrophages showed increased expression of IFN-β in response to direct irradiation, or to treatment with irradiated cancer cells, further supporting our hypothesis. We then tested the efficiency of a single dose RT in three subcutaneous and one orthotopic tumor models. In all situations, myeloid deletion of Trim33 led to a significantly improved response after RT, leading to a complete and durable response in most of the treated mice bearing orthotopic oral tumors. This effect required the IFN-I pathway, and the presence of CD8+ T lymphocytes, but not NK cells. In addition, cured mice were capable of rejecting a secondary tumor challenge, demonstrating an in situ vaccination effect. We conclude that deleting Trim33 in myeloid cells improves RT efficiency, through a mechanism involving the IFN-I pathway and the immune response. Our work suggests that myeloid Trim33 is a host factor affecting the tumor response to RT, thus representing a new potential therapeutic target for modifying RT responses.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.1,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142342193","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
Hyper-Interferon Sensitive influenza induces adaptive immune responses and overcomes resistance to anti-PD-1 in murine non-small cell lung cancer. 超干扰素敏感性流感诱导适应性免疫反应,克服小鼠非小细胞肺癌对抗 PD-1 的耐药性。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-26 DOI: 10.1158/2326-6066.CIR-23-1075
Yushen Du, Ramin Salehi-Rad, Tian-Hao Zhang, William P Crosson, Jensen Abascal, Dongdong Chen, Yuan Shi, Hong Jiang, Yen-Wen Tseng, Xi Ma, Mengying Hong, Sihan Wang, Xijuan Wang, Kejun Tang, Shiyao Hu, Yuting Li, Shaokai Ni, Yiqi Cai, Shahed Tappuni, Yong Shen, Bin Liu, Ren Sun

Despite recent advances in immunotherapy with immune checkpoint inhibitors (ICI), many patients with non-small cell lung cancer (NSCLC) fail to respond or develop resistance after an initial response. In situ vaccination (ISV) with engineered viruses has emerged as a promising antigen-agnostic strategy that can both condition the tumor microenvironment (TME) and augment anti-tumor T cell responses to overcome immune resistance. We engineered a live attenuated viral vaccine, Hyper-Interferon Sensitive virus (HIS), by conducting a genome-wide functional screening and introducing eight interferon (IFN)-sensitive mutations in the influenza genome. Compared to wild-type (WT) influenza, HIS replication was attenuated in immunocompetent hosts, enhancing its potential as a safe option for cancer therapy. HIS ISV elicited robust yet transient type I IFN responses in murine NSCLCs, leading to an enrichment of polyfunctional effector Th1 CD4 and cytotoxic CD8 T cells into the tumor. HIS ISV demonstrated enhanced anti-tumor efficacy compared to WT in multiple syngeneic murine models of NSCLC with distinct driver mutations and varying mutational burden. This efficacy was dependent on host type 1 IFN responses and T lymphocytes. HIS ISV overcame resistance to anti-PD-1 in LKB-1 deficient murine NSCLC, resulting in improved overall survival and enduring systemic tumor-specific immunity. These studies provide compelling evidence to support further clinical evaluation of HIS as a novel 'off-the-shelf' ISV strategy for patients with NSCLC refractory to ICI.

尽管免疫检查点抑制剂(ICI)的免疫疗法取得了最新进展,但许多非小细胞肺癌(NSCLC)患者在初次应答后仍无反应或产生抗药性。使用工程病毒进行原位接种(ISV)已成为一种很有前景的抗原诊断策略,它既能调节肿瘤微环境(TME),又能增强抗肿瘤T细胞反应,从而克服免疫耐受。我们通过进行全基因组功能筛选并在流感基因组中引入八个干扰素(IFN)敏感突变,设计出了一种减毒活疫苗--高干扰素敏感病毒(HIS)。与野生型(WT)流感相比,HIS 在免疫功能健全的宿主体内的复制能力减弱,从而提高了其作为癌症治疗安全选择的潜力。HIS ISV能在小鼠NSCLCs中引起强健但短暂的I型IFN反应,导致肿瘤中富集多功能效应Th1 CD4和细胞毒性CD8 T细胞。与 WT 相比,HIS ISV 在多种具有不同驱动基因突变和不同突变负荷的 NSCLC 合成小鼠模型中显示出更强的抗肿瘤功效。这种疗效取决于宿主的 1 型 IFN 反应和 T 淋巴细胞。HIS ISV克服了LKB-1缺陷小鼠NSCLC对抗PD-1的耐药性,从而提高了总生存率和持久的全身肿瘤特异性免疫力。这些研究提供了令人信服的证据,支持进一步对 HIS 进行临床评估,将其作为 ICI 难治性 NSCLC 患者的一种新型 "现成 "ISV 策略。
{"title":"Hyper-Interferon Sensitive influenza induces adaptive immune responses and overcomes resistance to anti-PD-1 in murine non-small cell lung cancer.","authors":"Yushen Du, Ramin Salehi-Rad, Tian-Hao Zhang, William P Crosson, Jensen Abascal, Dongdong Chen, Yuan Shi, Hong Jiang, Yen-Wen Tseng, Xi Ma, Mengying Hong, Sihan Wang, Xijuan Wang, Kejun Tang, Shiyao Hu, Yuting Li, Shaokai Ni, Yiqi Cai, Shahed Tappuni, Yong Shen, Bin Liu, Ren Sun","doi":"10.1158/2326-6066.CIR-23-1075","DOIUrl":"10.1158/2326-6066.CIR-23-1075","url":null,"abstract":"<p><p>Despite recent advances in immunotherapy with immune checkpoint inhibitors (ICI), many patients with non-small cell lung cancer (NSCLC) fail to respond or develop resistance after an initial response. In situ vaccination (ISV) with engineered viruses has emerged as a promising antigen-agnostic strategy that can both condition the tumor microenvironment (TME) and augment anti-tumor T cell responses to overcome immune resistance. We engineered a live attenuated viral vaccine, Hyper-Interferon Sensitive virus (HIS), by conducting a genome-wide functional screening and introducing eight interferon (IFN)-sensitive mutations in the influenza genome. Compared to wild-type (WT) influenza, HIS replication was attenuated in immunocompetent hosts, enhancing its potential as a safe option for cancer therapy. HIS ISV elicited robust yet transient type I IFN responses in murine NSCLCs, leading to an enrichment of polyfunctional effector Th1 CD4 and cytotoxic CD8 T cells into the tumor. HIS ISV demonstrated enhanced anti-tumor efficacy compared to WT in multiple syngeneic murine models of NSCLC with distinct driver mutations and varying mutational burden. This efficacy was dependent on host type 1 IFN responses and T lymphocytes. HIS ISV overcame resistance to anti-PD-1 in LKB-1 deficient murine NSCLC, resulting in improved overall survival and enduring systemic tumor-specific immunity. These studies provide compelling evidence to support further clinical evaluation of HIS as a novel 'off-the-shelf' ISV strategy for patients with NSCLC refractory to ICI.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.1,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142342194","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
Targeting BTN2A1 enhances Vγ9Vδ2 T-cell effector functions and triggers tumor cell pyroptosis. 靶向 BTN2A1 可增强 Vγ9Vδ2 T 细胞效应器功能并引发肿瘤细胞热解。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-20 DOI: 10.1158/2326-6066.CIR-23-0868
Anne-Charlotte Le Floch, Caroline Imbert, Nicolas Boucherit, Laurent Gorvel, Stéphane Fattori, Florence Orlanducci, Aude Le Roy, Lorenzo Archetti, Lydie Crescence, Laurence Panicot-Dubois, Christophe Dubois, Norbert Vey, Antoine Briantais, Amandine Anastasio, Carla Cano, Geoffrey Guittard, Mathieu Frechin, Daniel Olive

Vγ9Vδ2 T cells are potent but elusive cytotoxic effectors. Butyrophilin subfamily 2 member A1 (BTN2A1) is a surface protein that has recently been shown to bind the Vγ9 chain of the γδ T-cell receptor (TCR) but its precise role in modulating Vγ9Vδ2 T-cell functions remains unknown. Here, we show that 107G3B5, a monoclonal BTN2A1 agonist antibody, was able to significantly enhance Vγ9Vδ2 T-cell functions against hematological or solid cell lines and against primary cells from adult acute lymphoblastic leukemia patients. New computer vision strategies applied to holotomographic microscopy videos showed that 107G3B5 enhanced the interaction between Vγ9Vδ2 T cells and target cells in a quantitative and qualitative manner. In addition, we found that Vγ9Vδ2 T cells activated by 107G3B5 induced caspase 3/7 activation in tumor cells, thereby triggering tumor cell death by pyroptosis. Together, these data demonstrate that targeting BTN2A1 with 107G3B5 enhances the Vγ9Vδ2 T-cell antitumor response by triggering the pyroptosis-induced immunogenic cell death. These new pyroptosis-based therapies have great potential to stimulate the immune system to fight cancer, especially "cold" tumors.

Vγ9Vδ2 T细胞是一种强大但难以捉摸的细胞毒性效应因子。嗜丁蛋白亚家族 2 成员 A1(BTN2A1)是一种表面蛋白,最近已被证明能结合γδ T 细胞受体(TCR)的 Vγ9 链,但它在调节 Vγ9Vδ2 T 细胞功能方面的确切作用仍不清楚。在这里,我们发现单克隆BTN2A1激动剂抗体107G3B5能显著增强Vγ9Vδ2 T细胞对血液或实体细胞系以及成人急性淋巴细胞白血病患者原代细胞的功能。将新的计算机视觉策略应用于全图显微镜视频显示,107G3B5以定量和定性的方式增强了Vγ9Vδ2 T细胞与靶细胞之间的相互作用。此外,我们还发现,107G3B5激活的Vγ9Vδ2 T细胞可诱导肿瘤细胞中的caspase 3/7活化,从而引发肿瘤细胞热解死亡。总之,这些数据表明,用 107G3B5 靶向 BTN2A1 可通过引发热解诱导的免疫原性细胞死亡来增强 Vγ9Vδ2 T 细胞的抗肿瘤反应。这些基于热蛋白沉积的新疗法在刺激免疫系统抗击癌症,尤其是 "冷 "肿瘤方面具有巨大潜力。
{"title":"Targeting BTN2A1 enhances Vγ9Vδ2 T-cell effector functions and triggers tumor cell pyroptosis.","authors":"Anne-Charlotte Le Floch, Caroline Imbert, Nicolas Boucherit, Laurent Gorvel, Stéphane Fattori, Florence Orlanducci, Aude Le Roy, Lorenzo Archetti, Lydie Crescence, Laurence Panicot-Dubois, Christophe Dubois, Norbert Vey, Antoine Briantais, Amandine Anastasio, Carla Cano, Geoffrey Guittard, Mathieu Frechin, Daniel Olive","doi":"10.1158/2326-6066.CIR-23-0868","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-23-0868","url":null,"abstract":"<p><p>Vγ9Vδ2 T cells are potent but elusive cytotoxic effectors. Butyrophilin subfamily 2 member A1 (BTN2A1) is a surface protein that has recently been shown to bind the Vγ9 chain of the γδ T-cell receptor (TCR) but its precise role in modulating Vγ9Vδ2 T-cell functions remains unknown. Here, we show that 107G3B5, a monoclonal BTN2A1 agonist antibody, was able to significantly enhance Vγ9Vδ2 T-cell functions against hematological or solid cell lines and against primary cells from adult acute lymphoblastic leukemia patients. New computer vision strategies applied to holotomographic microscopy videos showed that 107G3B5 enhanced the interaction between Vγ9Vδ2 T cells and target cells in a quantitative and qualitative manner. In addition, we found that Vγ9Vδ2 T cells activated by 107G3B5 induced caspase 3/7 activation in tumor cells, thereby triggering tumor cell death by pyroptosis. Together, these data demonstrate that targeting BTN2A1 with 107G3B5 enhances the Vγ9Vδ2 T-cell antitumor response by triggering the pyroptosis-induced immunogenic cell death. These new pyroptosis-based therapies have great potential to stimulate the immune system to fight cancer, especially \"cold\" tumors.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.1,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142280528","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
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