首页 > 最新文献

Cancer immunology research最新文献

英文 中文
Intermittent MEK inhibition with GITR co-stimulation rescues T-cell function for increased efficacy with CTLA-4 blockade in solid tumor models. 间歇性 MEK 抑制与 GITR 协同刺激可挽救 T 细胞功能,从而提高 CTLA-4 阻断剂在实体瘤模型中的疗效。
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-06-18 DOI: 10.1158/2326-6066.CIR-23-0729
Lauren Dong, Hyejin Choi, Sadna Budhu, Isabell Schulze, Svena Verma, Levi Mark Mangarin, Valeria Estrada Navarro, Nezar Mehanna, Jonathan F Khan, Divya Venkatesh, Daniel Thach, Neal Rosen, Jedd D Wolchok, Taha Merghoub

MEK inhibitors (MEKis) have shown limited success as a treatment for MAPK/ERK pathway-dependent cancers due to various resistance mechanisms tumor cells can employ. CH5126766 (CKI27) is an inhibitor that binds to MEK and prevents release of RAF, reducing the relief of negative feedback commonly observed with other MEKis. We observed that CKI27 increased MHC expression on tumor cells and improved T cell-mediated killing. Yet, CKI27 also decreased T-cell proliferation, activation, and cytolytic activity by inhibiting the MAPK/ERK pathway that is activated downstream of T cell-receptor signaling. Therefore, we aimed to balance the positive and negative immunomodulatory effects of MEKis for optimal combination with immunotherapy. Intermittent administration of CKI27 allowed T cells to partially recover and co-stimulation via GITR and OX-40 agonist antibodies completely alleviated inhibition of function. In Kras mutant lung and colon tumor mouse models, intermittent CKI27 and anti-GITR significantly decreased tumor growth and prolonged survival when further combined with CTLA-4 immune checkpoint blockade. Moreover, this triple combination increased CD8+ and CD4+ T-cell proliferation, activation, and effector/memory subsets in the tumor draining lymph nodes and tumors and led to intratumoral regulatory T cell (Treg) destabilization. These data, collectively, will allow for more informed decisions when optimizing combination regimens by overcoming resistance, reducing toxicity, and generating long-term immune responses.

MEK 抑制剂(MEKis)在治疗依赖 MAPK/ERK 通路的癌症方面效果有限,原因是肿瘤细胞会采用各种抗药机制。CH5126766 (CKI27)是一种与MEK结合的抑制剂,它能阻止RAF的释放,减少其他MEK抑制剂常见的负反馈。我们观察到,CKI27 增加了肿瘤细胞上的 MHC 表达,提高了 T 细胞介导的杀伤力。然而,CKI27 也通过抑制 T 细胞受体信号传导下游激活的 MAPK/ERK 通路,降低了 T 细胞的增殖、活化和细胞溶解活性。因此,我们的目标是平衡 MEKis 的正负免疫调节作用,以实现与免疫疗法的最佳结合。间歇性给药 CKI27 可使 T 细胞部分恢复,而通过 GITR 和 OX-40 激动剂抗体进行联合刺激可完全缓解对 T 细胞功能的抑制。在 Kras 突变的肺癌和结肠癌小鼠模型中,间歇性服用 CKI27 和抗 GITR 能显著降低肿瘤生长,并延长存活时间,如果进一步结合 CTLA-4 免疫检查点阻断疗法。此外,这种三联疗法增加了肿瘤引流淋巴结和肿瘤中 CD8+ 和 CD4+ T 细胞的增殖、活化和效应/记忆亚群,并导致瘤内调节性 T 细胞(Treg)不稳定。这些数据将有助于在优化联合疗法时做出更明智的决定,克服耐药性、降低毒性并产生长期免疫反应。
{"title":"Intermittent MEK inhibition with GITR co-stimulation rescues T-cell function for increased efficacy with CTLA-4 blockade in solid tumor models.","authors":"Lauren Dong, Hyejin Choi, Sadna Budhu, Isabell Schulze, Svena Verma, Levi Mark Mangarin, Valeria Estrada Navarro, Nezar Mehanna, Jonathan F Khan, Divya Venkatesh, Daniel Thach, Neal Rosen, Jedd D Wolchok, Taha Merghoub","doi":"10.1158/2326-6066.CIR-23-0729","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-23-0729","url":null,"abstract":"<p><p>MEK inhibitors (MEKis) have shown limited success as a treatment for MAPK/ERK pathway-dependent cancers due to various resistance mechanisms tumor cells can employ. CH5126766 (CKI27) is an inhibitor that binds to MEK and prevents release of RAF, reducing the relief of negative feedback commonly observed with other MEKis. We observed that CKI27 increased MHC expression on tumor cells and improved T cell-mediated killing. Yet, CKI27 also decreased T-cell proliferation, activation, and cytolytic activity by inhibiting the MAPK/ERK pathway that is activated downstream of T cell-receptor signaling. Therefore, we aimed to balance the positive and negative immunomodulatory effects of MEKis for optimal combination with immunotherapy. Intermittent administration of CKI27 allowed T cells to partially recover and co-stimulation via GITR and OX-40 agonist antibodies completely alleviated inhibition of function. In Kras mutant lung and colon tumor mouse models, intermittent CKI27 and anti-GITR significantly decreased tumor growth and prolonged survival when further combined with CTLA-4 immune checkpoint blockade. Moreover, this triple combination increased CD8+ and CD4+ T-cell proliferation, activation, and effector/memory subsets in the tumor draining lymph nodes and tumors and led to intratumoral regulatory T cell (Treg) destabilization. These data, collectively, will allow for more informed decisions when optimizing combination regimens by overcoming resistance, reducing toxicity, and generating long-term immune responses.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141417944","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
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 10.1 1区 医学 Q1 Medicine Pub Date : 2024-06-18 DOI: 10.1158/2326-6066.CIR-23-0701
Daniel Peters, Lena M Kranz, David Eisel, Mustafa Diken, Sebastian Kreiter, Özlem Tureci, Ugur Sahin, Mathias Vormehr

Interleukin-2 (IL-2) is a crucial cytokine in T-cell immunity and a promising combination partner to boost cancer vaccine efficacy. However, therapeutic application of IL-2 is hampered by its short half-life and substantial toxicities. Herein, we report preclinical characterization of a mouse serum albumin-IL-2 fusion protein (Alb-IL2) encoded on nucleoside-modified RNA 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 two 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 to murine recombinant IL-2 (rIL-2). In combination with RNA-LPX vaccines, Alb-IL2 RNA-NP administration highly increased 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 Tureci, Ugur Sahin, Mathias Vormehr","doi":"10.1158/2326-6066.CIR-23-0701","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-23-0701","url":null,"abstract":"<p><p>Interleukin-2 (IL-2) is a crucial cytokine in T-cell immunity and a promising combination partner to boost cancer vaccine efficacy. However, therapeutic application of IL-2 is hampered by its short half-life and substantial toxicities. Herein, we report preclinical characterization of a mouse serum albumin-IL-2 fusion protein (Alb-IL2) encoded on nucleoside-modified RNA 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 two 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 to murine recombinant IL-2 (rIL-2). In combination with RNA-LPX vaccines, Alb-IL2 RNA-NP administration highly increased 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":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-06-18","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
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 Medicine 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":null,"pages":null},"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
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":null,"pages":null},"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":null,"pages":null},"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
Intratumoral TREX1 Induction Promotes Immune Evasion by Limiting Type I IFN. 瘤内TREX1诱导通过限制I型干扰素促进免疫逃避。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-23-1093
Eléonore Toufektchan, Alexandra Dananberg, Josefine Striepen, James H Hickling, Abraham Shim, Yanyang Chen, Ashley Nichols, Mercedes A Duran Paez, Lisa Mohr, Samuel F Bakhoum, John Maciejowski

Chromosomal instability is a hallmark of human cancer that is associated with aggressive disease characteristics. Chromosome mis-segregations help fuel natural selection, but they risk provoking a cGAS-STING immune response through the accumulation of cytosolic DNA. The mechanisms of how tumors benefit from chromosomal instability while mitigating associated risks, such as enhanced immune surveillance, are poorly understood. Here, we identify cGAS-STING-dependent upregulation of the nuclease TREX1 as an adaptive, negative feedback mechanism that promotes immune evasion through digestion of cytosolic DNA. TREX1 loss diminishes tumor growth, prolongs survival of host animals, increases tumor immune infiltration, and potentiates response to immune checkpoint blockade selectively in tumors capable of mounting a type I IFN response downstream of STING. Together, these data demonstrate that TREX1 induction shields chromosomally unstable tumors from immune surveillance by dampening type I IFN production and suggest that TREX1 inhibitors might be used to selectively target tumors that have retained the inherent ability to mount an IFN response downstream of STING. See related article by Lim et al., p. 663.

染色体不稳定性是人类癌症的一个特征,与侵袭性疾病特征有关。染色体错误分离有助于促进自然选择,但它们有可能通过细胞膜 DNA 的积累引发 cGAS-STING 免疫反应。人们对肿瘤如何从染色体不稳定性中获益,同时减轻相关风险(如增强免疫监视)的机制知之甚少。在这里,我们发现 cGAS-STING 依赖性上调核酸酶 TREX1 是一种适应性负反馈机制,可通过消化细胞膜 DNA 促进免疫逃避。TREX1 的缺失会减少肿瘤的生长,延长宿主动物的存活时间,增加肿瘤的免疫浸润,并有选择性地增强肿瘤对免疫检查点阻断的反应,这些肿瘤能够在 STING 下游产生 I 型干扰素反应。这些数据共同证明,TREX1诱导通过抑制I型干扰素的产生,使染色体不稳定的肿瘤免受免疫监视,并表明TREX1抑制剂可用于选择性地靶向保留了在STING下游产生干扰素反应的固有能力的肿瘤。
{"title":"Intratumoral TREX1 Induction Promotes Immune Evasion by Limiting Type I IFN.","authors":"Eléonore Toufektchan, Alexandra Dananberg, Josefine Striepen, James H Hickling, Abraham Shim, Yanyang Chen, Ashley Nichols, Mercedes A Duran Paez, Lisa Mohr, Samuel F Bakhoum, John Maciejowski","doi":"10.1158/2326-6066.CIR-23-1093","DOIUrl":"10.1158/2326-6066.CIR-23-1093","url":null,"abstract":"<p><p>Chromosomal instability is a hallmark of human cancer that is associated with aggressive disease characteristics. Chromosome mis-segregations help fuel natural selection, but they risk provoking a cGAS-STING immune response through the accumulation of cytosolic DNA. The mechanisms of how tumors benefit from chromosomal instability while mitigating associated risks, such as enhanced immune surveillance, are poorly understood. Here, we identify cGAS-STING-dependent upregulation of the nuclease TREX1 as an adaptive, negative feedback mechanism that promotes immune evasion through digestion of cytosolic DNA. TREX1 loss diminishes tumor growth, prolongs survival of host animals, increases tumor immune infiltration, and potentiates response to immune checkpoint blockade selectively in tumors capable of mounting a type I IFN response downstream of STING. Together, these data demonstrate that TREX1 induction shields chromosomally unstable tumors from immune surveillance by dampening type I IFN production and suggest that TREX1 inhibitors might be used to selectively target tumors that have retained the inherent ability to mount an IFN response downstream of STING. See related article by Lim et al., p. 663.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11148545/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139971036","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 Sampling of Highlights from the Literature. 文献精华选集》。
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-12-6-WWR
{"title":"A Sampling of Highlights from the Literature.","authors":"","doi":"10.1158/2326-6066.CIR-12-6-WWR","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-12-6-WWR","url":null,"abstract":"","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141236956","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
Randomized Phase II Study Evaluating the Addition of Pembrolizumab to Radium-223 in Metastatic Castration-resistant Prostate Cancer. 评估转移性钙化抗性前列腺癌患者在镭-223基础上加用 Pembrolizumab 的随机 II 期研究。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-22-0306
Atish D Choudhury, Lucia Kwak, Alexander Cheung, Kathryn M Allaire, Jaqueline Marquez, David D Yang, Abhishek Tripathi, Jacqueline M Kilar, Meredith Flynn, Brianna Maynard, Rebecca Reichel, Amanda F Pace, Brandon K Chen, Eliezer M Van Allen, Kerry Kilbridge, Xiao X Wei, Bradley A McGregor, Mark M Pomerantz, Rupal S Bhatt, Christopher J Sweeney, Glenn J Bubley, Heather A Jacene, Mary-Ellen Taplin, Franklin W Huang, Lauren C Harshman, Lawrence Fong

The checkpoint immunotherapeutic pembrolizumab induces responses in a small minority of patients with metastatic castration-resistant prostate cancer (mCRPC). Radium-223 (R223) may increase immunogenicity of bone metastases and increase pembrolizumab (P) activity. In a randomized phase II study, we assessed the effect of R223+P compared with R223 on tumor immune infiltration, safety, and clinical outcomes in patients with mCRPC. The primary endpoint was differences in CD4+ and CD8+ T-cell infiltrate in 8-week versus baseline bone metastasis biopsies; secondary endpoints were safety, radiographic progression-free survival (rPFS), and overall survival (OS). Of the 42 treated patients (29 R223+P, 13 R223), 18 R223+P and 8 R223 patients had evaluable paired tumor biopsies. Median fold-change of CD4+ T cells was -0.7 (range: -9.3 to 4.7) with R223+P and 0.1 (-11.1 to 3.7) with R223 (P = 0.66); for CD8+ T cells, median fold-change was -0.6 (-7.4 to 5.3) with R223+P and -1.3 (-3.1 to 4.8) with R223 (P = 0.66). Median rPFS and OS was 6.1 (95% confidence interval: 2.7-11.0) and 16.9 months [12.7-not reached (NR)], respectively, with R223+P and 5.7 (2.6-NR) and 16.0 (9.0-NR), respectively, with R223. Although R223+P was well tolerated with no unexpected toxicity, the combination did not improve efficacy. High-dimensional flow cytometry demonstrated minimal immune modulation with R223, whereas R223+P induced CTLA-4 expression on circulating CD4+ T cells. Clinical responders possessed lower circulating frequencies of Ki67+ T and myeloid cells at baseline and higher circulating frequencies of TIM-3+ T and myeloid cells by week 9. Although R223+P did not induce T-cell infiltration into the tumor microenvironment, exhaustion of induced peripheral T-cell immune responses may dampen the combination's clinical activity.

检查点免疫疗法 pembrolizumab 可诱导少数转移性去势抵抗性前列腺癌(mCRPC)患者产生反应。镭-223(R223)可能会增加骨转移灶的免疫原性并提高pembrolizumab(P)的活性。在一项随机II期研究中,我们评估了R223+P与R223相比对mCRPC患者肿瘤免疫浸润、安全性和临床结果的影响。研究的主要终点是 8 周骨转移活检中 CD4+ 和 CD8+ T 细胞浸润与基线的差异;次要终点是安全性、放射学无进展生存期 (rPFS) 和总生存期 (OS)。在42例接受治疗的患者(29例R223+P,13例R223)中,18例R223+P和8例R223患者有可评估的配对肿瘤活检。R223+P的CD4+ T细胞中位折叠变化为-0.7(范围:-9.3至4.7),R223的CD4+ T细胞中位折叠变化为0.1(-11.1至3.7)(P = 0.66);R223+P的CD8+ T细胞中位折叠变化为-0.6(-7.4至5.3),R223的CD8+ T细胞中位折叠变化为-1.3(-3.1至4.8)(P = 0.66)。R223+P的中位rPFS和OS分别为6.1个月(95%置信区间:2.7-11.0)和16.9个月[12.7个月未达标(NR)],R223的中位rPFS和OS分别为5.7个月(2.6-NR)和16.0个月(9.0-NR)。虽然 R223+P 的耐受性良好,没有出现意外毒性,但联合用药并未提高疗效。高维流式细胞术显示,R223 对免疫调节的作用微乎其微,而 R223+P 则诱导循环 CD4+ T 细胞表达 CTLA-4。临床应答者在基线时拥有较低的 Ki67+ T 细胞和髓系细胞循环频率,到第 9 周时拥有较高的 TIM-3+ T 细胞和髓系细胞循环频率。虽然R223+P没有诱导T细胞浸润到肿瘤微环境中,但诱导的外周T细胞免疫反应耗竭可能会抑制该组合的临床活性。
{"title":"Randomized Phase II Study Evaluating the Addition of Pembrolizumab to Radium-223 in Metastatic Castration-resistant Prostate Cancer.","authors":"Atish D Choudhury, Lucia Kwak, Alexander Cheung, Kathryn M Allaire, Jaqueline Marquez, David D Yang, Abhishek Tripathi, Jacqueline M Kilar, Meredith Flynn, Brianna Maynard, Rebecca Reichel, Amanda F Pace, Brandon K Chen, Eliezer M Van Allen, Kerry Kilbridge, Xiao X Wei, Bradley A McGregor, Mark M Pomerantz, Rupal S Bhatt, Christopher J Sweeney, Glenn J Bubley, Heather A Jacene, Mary-Ellen Taplin, Franklin W Huang, Lauren C Harshman, Lawrence Fong","doi":"10.1158/2326-6066.CIR-22-0306","DOIUrl":"10.1158/2326-6066.CIR-22-0306","url":null,"abstract":"<p><p>The checkpoint immunotherapeutic pembrolizumab induces responses in a small minority of patients with metastatic castration-resistant prostate cancer (mCRPC). Radium-223 (R223) may increase immunogenicity of bone metastases and increase pembrolizumab (P) activity. In a randomized phase II study, we assessed the effect of R223+P compared with R223 on tumor immune infiltration, safety, and clinical outcomes in patients with mCRPC. The primary endpoint was differences in CD4+ and CD8+ T-cell infiltrate in 8-week versus baseline bone metastasis biopsies; secondary endpoints were safety, radiographic progression-free survival (rPFS), and overall survival (OS). Of the 42 treated patients (29 R223+P, 13 R223), 18 R223+P and 8 R223 patients had evaluable paired tumor biopsies. Median fold-change of CD4+ T cells was -0.7 (range: -9.3 to 4.7) with R223+P and 0.1 (-11.1 to 3.7) with R223 (P = 0.66); for CD8+ T cells, median fold-change was -0.6 (-7.4 to 5.3) with R223+P and -1.3 (-3.1 to 4.8) with R223 (P = 0.66). Median rPFS and OS was 6.1 (95% confidence interval: 2.7-11.0) and 16.9 months [12.7-not reached (NR)], respectively, with R223+P and 5.7 (2.6-NR) and 16.0 (9.0-NR), respectively, with R223. Although R223+P was well tolerated with no unexpected toxicity, the combination did not improve efficacy. High-dimensional flow cytometry demonstrated minimal immune modulation with R223, whereas R223+P induced CTLA-4 expression on circulating CD4+ T cells. Clinical responders possessed lower circulating frequencies of Ki67+ T and myeloid cells at baseline and higher circulating frequencies of TIM-3+ T and myeloid cells by week 9. Although R223+P did not induce T-cell infiltration into the tumor microenvironment, exhaustion of induced peripheral T-cell immune responses may dampen the combination's clinical activity.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11148544/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140326365","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
An IRF2-Expressing Oncolytic Virus Changes the Susceptibility of Tumor Cells to Antitumor T Cells and Promotes Tumor Clearance. 表达 IRF2 的溶瘤病毒可改变肿瘤细胞对抗肿瘤 T 细胞的易感性并促进肿瘤清除
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-23-0573
Lulu Shao, Rashmi Srivastava, Greg M Delgoffe, Stephen H Thorne, Saumendra N Sarkar

IFN regulatory factor 1 (IRF1) can promote antitumor immunity. However, we have shown previously that in the tumor cell, IRF1 can promote tumor growth, and IRF1-deficient tumor cells exhibit severely restricted tumor growth in several syngeneic mouse tumor models. Here, we investigate the potential of functionally modulating IRF1 to reduce tumor progression and prolong survival. Using inducible IRF1 expression, we established that it is possible to regulate IRF1 expression to modulate tumor progression in established B16-F10 tumors. Expression of IRF2, which is a functional antagonist of IRF1, downregulated IFNγ-induced expression of inhibitory ligands, upregulated MHC-related molecules, and slowed tumor growth and extended survival. We characterized the functional domain(s) of IRF2 needed for this antitumor activity, showing that a full-length IRF2 was required for its antitumor functions. Finally, using an oncolytic vaccinia virus as a delivery platform, we showed that IRF2-expressing vaccinia virus suppressed tumor progression and prolonged survival in multiple tumor models. These results suggest the potency of targeting IRF1 and using IRF2 to modulate immunotherapy.

干扰素调节因子 1(IRF1)可促进抗肿瘤免疫。然而,我们之前已经证明,在肿瘤细胞中,IRF1 可以促进肿瘤生长,在几种合成小鼠肿瘤模型中,IRF1 缺失的肿瘤细胞表现出严重的肿瘤生长受限。在这里,我们研究了从功能上调节 IRF1 以减少肿瘤进展和延长生存期的可能性。通过诱导 IRF1 的表达,我们发现可以通过调节 IRF1 的表达来调控 B16-F10 肿瘤的进展。IRF2是IRF1的功能性拮抗剂,它的表达能下调IFN 诱导的抑制性配体的表达,上调MHC相关分子,减缓肿瘤生长并延长生存期。我们对这种抗肿瘤活性所需的IRF2功能域进行了鉴定,结果表明其抗肿瘤功能需要全长的IRF2。最后,我们利用溶瘤疫苗病毒作为递送平台,在多种肿瘤模型中证明了表达 IRF2 的疫苗病毒能抑制肿瘤进展并延长生存期。这些结果表明了靶向 IRF1 和利用 IRF2 调节免疫疗法的有效性。
{"title":"An IRF2-Expressing Oncolytic Virus Changes the Susceptibility of Tumor Cells to Antitumor T Cells and Promotes Tumor Clearance.","authors":"Lulu Shao, Rashmi Srivastava, Greg M Delgoffe, Stephen H Thorne, Saumendra N Sarkar","doi":"10.1158/2326-6066.CIR-23-0573","DOIUrl":"10.1158/2326-6066.CIR-23-0573","url":null,"abstract":"<p><p>IFN regulatory factor 1 (IRF1) can promote antitumor immunity. However, we have shown previously that in the tumor cell, IRF1 can promote tumor growth, and IRF1-deficient tumor cells exhibit severely restricted tumor growth in several syngeneic mouse tumor models. Here, we investigate the potential of functionally modulating IRF1 to reduce tumor progression and prolong survival. Using inducible IRF1 expression, we established that it is possible to regulate IRF1 expression to modulate tumor progression in established B16-F10 tumors. Expression of IRF2, which is a functional antagonist of IRF1, downregulated IFNγ-induced expression of inhibitory ligands, upregulated MHC-related molecules, and slowed tumor growth and extended survival. We characterized the functional domain(s) of IRF2 needed for this antitumor activity, showing that a full-length IRF2 was required for its antitumor functions. Finally, using an oncolytic vaccinia virus as a delivery platform, we showed that IRF2-expressing vaccinia virus suppressed tumor progression and prolonged survival in multiple tumor models. These results suggest the potency of targeting IRF1 and using IRF2 to modulate immunotherapy.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11150089/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140183844","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
Metabolic reprogramming of tumor-associated macrophages using glutamine antagonist JHU083 drives tumor immunity in myeloid-rich prostate and bladder cancer tumors 利用谷氨酰胺拮抗剂 JHU083 对肿瘤相关巨噬细胞进行代谢重编程,促进富含骨髓的前列腺癌和膀胱癌肿瘤的肿瘤免疫功能
IF 10.1 1区 医学 Q1 Medicine Pub Date : 2024-05-03 DOI: 10.1158/2326-6066.cir-23-1105
Monali Praharaj, Fan Shen, Alex J. Lee, Liang Zhao, Thomas R. Nirschl, Debebe Theodros, Alok K. Singh, Xiaoxu Wang, Kenneth M. Adusei, Kara A. Lombardo, Raekwon A. Williams, Laura A. Sena, Elizabeth A. Thompson, Ada Tam, Srinivasan Yegnasubramanian, Edward J. Pearce, Robert D. Leone, Jesse Alt, Rana Rais, Barbara S. Slusher, Drew M. Pardoll, Jonathan D. Powell, Jelani C. Zarif
Glutamine metabolism in tumor microenvironments critically regulates anti-tumor immunity. Using glutamine-antagonist prodrug JHU083, we report potent tumor growth inhibition in urologic tumors by JHU083-reprogrammed tumor-associated macrophages (TAMs) and tumor-infiltrating monocytes (TIMs). We show JHU083-mediated glutamine antagonism in tumor microenvironments induces TNF, pro-inflammatory, and mTORC1 signaling in intratumoral TAM clusters. JHU083-reprogrammed TAMs also exhibit increased tumor cell phagocytosis and diminished pro-angiogenic capacities. In vivo inhibition of TAM glutamine consumption resulted in increased glycolysis, a broken TCA cycle, and purine metabolism disruption. Although the anti-tumor effect of glutamine antagonism on tumor-infiltrating T cells was moderate, JHU083 promoted a stem cell-like phenotype in CD8+ T cells and decreased Treg abundance. Finally, JHU083 caused a ubiquitous shutdown in glutamine utilizing metabolic pathways in tumor cells, leading to reduced HIF-1alpha, c-MYC phosphorylation, and induction of tumor cell apoptosis, all key anti-tumor features.
肿瘤微环境中的谷氨酰胺代谢对抗肿瘤免疫起着至关重要的调节作用。利用谷氨酰胺拮抗剂原药 JHU083,我们报告了 JHU083 重编程的肿瘤相关巨噬细胞(TAMs)和肿瘤浸润单核细胞(TIMs)对泌尿系统肿瘤生长的强效抑制作用。我们发现,JHU083 在肿瘤微环境中介导的谷氨酰胺拮抗作用会诱导瘤内 TAM 集群中的 TNF、促炎和 mTORC1 信号转导。经过 JHU083 重编程的 TAM 还表现出更强的肿瘤细胞吞噬能力和更弱的促血管生成能力。体内抑制 TAM 谷氨酰胺消耗会导致糖酵解增加、TCA 循环中断和嘌呤代谢紊乱。虽然谷氨酰胺拮抗剂对肿瘤浸润T细胞的抗肿瘤效果一般,但JHU083促进了CD8+ T细胞的干细胞样表型,并降低了Treg的丰度。最后,JHU083 导致肿瘤细胞中谷氨酰胺利用代谢途径的普遍关闭,从而降低了 HIF-1alpha、c-MYC 磷酸化,并诱导肿瘤细胞凋亡,这些都是抗肿瘤的关键特征。
{"title":"Metabolic reprogramming of tumor-associated macrophages using glutamine antagonist JHU083 drives tumor immunity in myeloid-rich prostate and bladder cancer tumors","authors":"Monali Praharaj, Fan Shen, Alex J. Lee, Liang Zhao, Thomas R. Nirschl, Debebe Theodros, Alok K. Singh, Xiaoxu Wang, Kenneth M. Adusei, Kara A. Lombardo, Raekwon A. Williams, Laura A. Sena, Elizabeth A. Thompson, Ada Tam, Srinivasan Yegnasubramanian, Edward J. Pearce, Robert D. Leone, Jesse Alt, Rana Rais, Barbara S. Slusher, Drew M. Pardoll, Jonathan D. Powell, Jelani C. Zarif","doi":"10.1158/2326-6066.cir-23-1105","DOIUrl":"https://doi.org/10.1158/2326-6066.cir-23-1105","url":null,"abstract":"Glutamine metabolism in tumor microenvironments critically regulates anti-tumor immunity. Using glutamine-antagonist prodrug JHU083, we report potent tumor growth inhibition in urologic tumors by JHU083-reprogrammed tumor-associated macrophages (TAMs) and tumor-infiltrating monocytes (TIMs). We show JHU083-mediated glutamine antagonism in tumor microenvironments induces TNF, pro-inflammatory, and mTORC1 signaling in intratumoral TAM clusters. JHU083-reprogrammed TAMs also exhibit increased tumor cell phagocytosis and diminished pro-angiogenic capacities. In vivo inhibition of TAM glutamine consumption resulted in increased glycolysis, a broken TCA cycle, and purine metabolism disruption. Although the anti-tumor effect of glutamine antagonism on tumor-infiltrating T cells was moderate, JHU083 promoted a stem cell-like phenotype in CD8+ T cells and decreased Treg abundance. Finally, JHU083 caused a ubiquitous shutdown in glutamine utilizing metabolic pathways in tumor cells, leading to reduced HIF-1alpha, c-MYC phosphorylation, and induction of tumor cell apoptosis, all key anti-tumor features.","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-05-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140841852","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