首页 > 最新文献

Cancer immunology research最新文献

英文 中文
Pancreatic Epithelial IL17/IL17RA Signaling Drives B7-H4 Expression to Promote Tumorigenesis. 胰腺上皮细胞IL-17/IL-17RA信号驱动B7-H4的表达,从而促进肿瘤发生。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-23-0527
Susana Castro-Pando, Rian M Howell, Le Li, Marilina Mascaro, Erika Y Faraoni, Olivereen Le Roux, David Romanin, Virginia Tahan, Erick Riquelme, Yu Zhang, Jay K Kolls, James P Allison, Guillermina Lozano, Seyed J Moghaddam, Florencia McAllister

IL17 is required for the initiation and progression of pancreatic cancer, particularly in the context of inflammation, as previously shown by genetic and pharmacological approaches. However, the cellular compartment and downstream molecular mediators of IL17-mediated pancreatic tumorigenesis have not been fully identified. This study examined the cellular compartment required by generating transgenic animals with IL17 receptor A (IL17RA), which was genetically deleted from either the pancreatic epithelial compartment or the hematopoietic compartment via generation of IL17RA-deficient (IL17-RA-/-) bone marrow chimeras, in the context of embryonically activated or inducible Kras. Deletion of IL17RA from the pancreatic epithelial compartment, but not from hematopoietic compartment, resulted in delayed initiation and progression of premalignant lesions and increased infiltration of CD8+ cytotoxic T cells to the tumor microenvironment. Absence of IL17RA in the pancreatic compartment affected transcriptional profiles of epithelial cells, modulating stemness, and immunological pathways. B7-H4, a known inhibitor of T-cell activation encoded by the gene Vtcn1, was the checkpoint molecule most upregulated via IL17 early during pancreatic tumorigenesis, and its genetic deletion delayed the development of pancreatic premalignant lesions and reduced immunosuppression. Thus, our data reveal that pancreatic epithelial IL17RA promotes pancreatic tumorigenesis by reprogramming the immune pancreatic landscape, which is partially orchestrated by regulation of B7-H4. Our findings provide the foundation of the mechanisms triggered by IL17 to mediate pancreatic tumorigenesis and reveal the avenues for early pancreatic cancer immune interception. See related Spotlight by Lee and Pasca di Magliano, p. 1130.

遗传学和药理学方法表明,IL-17 是胰腺癌发生和发展的必要条件,尤其是在炎症背景下。IL-17 介导的胰腺肿瘤发生的细胞区系和下游分子介质尚未完全确定。我们在胚胎激活或诱导 Kras 的背景下,通过产生白细胞介素 17 受体 A(IL-17RA)基因被从胰腺上皮细胞区系中删除的转基因动物,以及通过产生 IL-17RA 缺失(IL17-RA-/-)骨髓嵌合体的造血细胞区系,对所需的细胞区系进行了研究。在胰腺上皮细胞中缺失IL-17RA,而在造血细胞中缺失IL-17RA,会导致恶性肿瘤前期病变的发生和发展延迟,并增加CD8+细胞毒性T细胞对肿瘤微环境的浸润。胰腺区系中缺少IL-17RA会影响上皮细胞的转录谱,从而调节干性和免疫途径。有趣的是,由Vtcn1基因编码的已知T细胞活化抑制剂B7-H4是胰腺肿瘤发生早期通过IL17上调最多的检查点分子,其基因缺失可延缓胰腺恶性肿瘤前病变的发展并减少免疫抑制。我们揭示了胰腺上皮IL-17RA对胰腺肿瘤发生的要求,它通过重编程胰腺免疫格局来实现,而胰腺免疫格局部分是由B7-H4调控的。
{"title":"Pancreatic Epithelial IL17/IL17RA Signaling Drives B7-H4 Expression to Promote Tumorigenesis.","authors":"Susana Castro-Pando, Rian M Howell, Le Li, Marilina Mascaro, Erika Y Faraoni, Olivereen Le Roux, David Romanin, Virginia Tahan, Erick Riquelme, Yu Zhang, Jay K Kolls, James P Allison, Guillermina Lozano, Seyed J Moghaddam, Florencia McAllister","doi":"10.1158/2326-6066.CIR-23-0527","DOIUrl":"10.1158/2326-6066.CIR-23-0527","url":null,"abstract":"<p><p>IL17 is required for the initiation and progression of pancreatic cancer, particularly in the context of inflammation, as previously shown by genetic and pharmacological approaches. However, the cellular compartment and downstream molecular mediators of IL17-mediated pancreatic tumorigenesis have not been fully identified. This study examined the cellular compartment required by generating transgenic animals with IL17 receptor A (IL17RA), which was genetically deleted from either the pancreatic epithelial compartment or the hematopoietic compartment via generation of IL17RA-deficient (IL17-RA-/-) bone marrow chimeras, in the context of embryonically activated or inducible Kras. Deletion of IL17RA from the pancreatic epithelial compartment, but not from hematopoietic compartment, resulted in delayed initiation and progression of premalignant lesions and increased infiltration of CD8+ cytotoxic T cells to the tumor microenvironment. Absence of IL17RA in the pancreatic compartment affected transcriptional profiles of epithelial cells, modulating stemness, and immunological pathways. B7-H4, a known inhibitor of T-cell activation encoded by the gene Vtcn1, was the checkpoint molecule most upregulated via IL17 early during pancreatic tumorigenesis, and its genetic deletion delayed the development of pancreatic premalignant lesions and reduced immunosuppression. Thus, our data reveal that pancreatic epithelial IL17RA promotes pancreatic tumorigenesis by reprogramming the immune pancreatic landscape, which is partially orchestrated by regulation of B7-H4. Our findings provide the foundation of the mechanisms triggered by IL17 to mediate pancreatic tumorigenesis and reveal the avenues for early pancreatic cancer immune interception. See related Spotlight by Lee and Pasca di Magliano, p. 1130.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11369627/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141261120","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
Blockade of IL1β and PD1 with Combination Chemotherapy Reduces Systemic Myeloid Suppression in Metastatic Pancreatic Cancer with Heterogeneous Effects in the Tumor. 通过联合化疗阻断IL-1β和PD-1可减少转移性胰腺癌的全身髓系抑制,并对肿瘤产生异质性影响。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-23-1073
Paul E Oberstein, Andressa Dias Costa, Emily A Kawaler, Victoire Cardot-Ruffino, Osama E Rahma, Nina Beri, Harshabad Singh, Thomas A Abrams, Leah H Biller, James M Cleary, Peter Enzinger, Brandon M Huffman, Nadine J McCleary, Kimberly J Perez, Douglas A Rubinson, Benjamin L Schlechter, Rishi Surana, Matthew B Yurgelun, S Jennifer Wang, Joshua Remland, Lauren K Brais, Naima Bollenrucher, Eugena Chang, Lestat R Ali, Patrick J Lenehan, Igor Dolgalev, Gregor Werba, Cibelle Lima, C Elizabeth Keheler, Keri M Sullivan, Michael Dougan, Cristina Hajdu, Maya Dajee, Marc R Pelletier, Saloney Nazeer, Matthew Squires, Dafna Bar-Sagi, Brian M Wolpin, Jonathan A Nowak, Diane M Simeone, Stephanie K Dougan

Innate inflammation promotes tumor development, although the role of innate inflammatory cytokines in established human tumors is unclear. Herein, we report clinical and translational results from a phase Ib trial testing whether IL1β blockade in human pancreatic cancer would alleviate myeloid immunosuppression and reveal antitumor T-cell responses to PD1 blockade. Patients with treatment-naïve advanced pancreatic ductal adenocarcinoma (n = 10) were treated with canakinumab, a high-affinity monoclonal human antiinterleukin-1β (IL1β), the PD1 blocking antibody spartalizumab, and gemcitabine/n(ab)paclitaxel. Analysis of paired peripheral blood from patients in the trial versus patients receiving multiagent chemotherapy showed a modest increase in HLA-DR+CD38+ activated CD8+ T cells and a decrease in circulating monocytic myeloid-derived suppressor cells (MDSC) by flow cytometry for patients in the trial but not in controls. Similarly, we used patient serum to differentiate monocytic MDSCs in vitro and showed that functional inhibition of T-cell proliferation was reduced when using on-treatment serum samples from patients in the trial but not when using serum from patients treated with chemotherapy alone. Within the tumor, we observed few changes in suppressive myeloid-cell populations or activated T cells as assessed by single-cell transcriptional profiling or multiplex immunofluorescence, although increases in CD8+ T cells suggest that improvements in the tumor immune microenvironment might be revealed by a larger study. Overall, the data indicate that exposure to PD1 and IL1β blockade induced a modest reactivation of peripheral CD8+ T cells and decreased circulating monocytic MDSCs; however, these changes did not lead to similarly uniform alterations in the tumor microenvironment.

先天性炎症会促进肿瘤的发展,但先天性炎症细胞因子在已确诊的人类肿瘤中的作用尚不清楚。在此,我们报告了一项Ib期试验的临床和转化结果,该试验测试了IL-1β在人类胰腺癌中的阻断作用是否会减轻髓系免疫抑制,并揭示了抗肿瘤T细胞对PD-1阻断作用的反应。采用高亲和力单克隆人类抗白细胞介素-1β(IL-1β)药物卡纳库单抗、PD-1阻断抗体spartalizumab和吉西他滨/n(ab)紫杉醇对治疗无效的晚期胰腺导管腺癌患者(10人)进行治疗。通过流式细胞术分析试验患者与接受多药化疗患者的配对外周血发现,试验患者的HLA-DR+CD38+活化CD8+ T细胞略有增加,循环中的单核细胞髓源性抑制细胞(MDSCs)有所减少,而对照组则没有。同样,我们使用患者血清在体外分化单核细胞MDSCs,结果表明,使用试验中患者的治疗中血清样本会减少对T细胞增殖的功能性抑制,而使用单独化疗患者的血清样本则不会。通过单细胞转录谱分析或多重免疫荧光评估,我们观察到肿瘤内抑制性髓系细胞群或活化的T细胞几乎没有发生变化,但CD8+ T细胞的增加表明,肿瘤免疫微环境的改善可能会通过更大规模的研究揭示出来。总之,这些数据表明,接触 PD-1 和 IL-1β 阻断剂会诱导外周 CD8+ T 细胞适度再激活,并减少循环中的单核细胞 MDSCs;然而,这些变化并没有导致肿瘤微环境发生类似的一致改变。
{"title":"Blockade of IL1β and PD1 with Combination Chemotherapy Reduces Systemic Myeloid Suppression in Metastatic Pancreatic Cancer with Heterogeneous Effects in the Tumor.","authors":"Paul E Oberstein, Andressa Dias Costa, Emily A Kawaler, Victoire Cardot-Ruffino, Osama E Rahma, Nina Beri, Harshabad Singh, Thomas A Abrams, Leah H Biller, James M Cleary, Peter Enzinger, Brandon M Huffman, Nadine J McCleary, Kimberly J Perez, Douglas A Rubinson, Benjamin L Schlechter, Rishi Surana, Matthew B Yurgelun, S Jennifer Wang, Joshua Remland, Lauren K Brais, Naima Bollenrucher, Eugena Chang, Lestat R Ali, Patrick J Lenehan, Igor Dolgalev, Gregor Werba, Cibelle Lima, C Elizabeth Keheler, Keri M Sullivan, Michael Dougan, Cristina Hajdu, Maya Dajee, Marc R Pelletier, Saloney Nazeer, Matthew Squires, Dafna Bar-Sagi, Brian M Wolpin, Jonathan A Nowak, Diane M Simeone, Stephanie K Dougan","doi":"10.1158/2326-6066.CIR-23-1073","DOIUrl":"10.1158/2326-6066.CIR-23-1073","url":null,"abstract":"<p><p>Innate inflammation promotes tumor development, although the role of innate inflammatory cytokines in established human tumors is unclear. Herein, we report clinical and translational results from a phase Ib trial testing whether IL1β blockade in human pancreatic cancer would alleviate myeloid immunosuppression and reveal antitumor T-cell responses to PD1 blockade. Patients with treatment-naïve advanced pancreatic ductal adenocarcinoma (n = 10) were treated with canakinumab, a high-affinity monoclonal human antiinterleukin-1β (IL1β), the PD1 blocking antibody spartalizumab, and gemcitabine/n(ab)paclitaxel. Analysis of paired peripheral blood from patients in the trial versus patients receiving multiagent chemotherapy showed a modest increase in HLA-DR+CD38+ activated CD8+ T cells and a decrease in circulating monocytic myeloid-derived suppressor cells (MDSC) by flow cytometry for patients in the trial but not in controls. Similarly, we used patient serum to differentiate monocytic MDSCs in vitro and showed that functional inhibition of T-cell proliferation was reduced when using on-treatment serum samples from patients in the trial but not when using serum from patients treated with chemotherapy alone. Within the tumor, we observed few changes in suppressive myeloid-cell populations or activated T cells as assessed by single-cell transcriptional profiling or multiplex immunofluorescence, although increases in CD8+ T cells suggest that improvements in the tumor immune microenvironment might be revealed by a larger study. Overall, the data indicate that exposure to PD1 and IL1β blockade induced a modest reactivation of peripheral CD8+ T cells and decreased circulating monocytic MDSCs; however, these changes did not lead to similarly uniform alterations in the tumor microenvironment.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11369625/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141579026","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
Nivolumab Reaches Brain Lesions in Patients with Recurrent Glioblastoma and Induces T-cell Activity and Upregulation of Checkpoint Pathways. Nivolumab可到达复发性胶质母细胞瘤患者的脑部病灶,诱导T细胞活性和检查点通路的上调。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-23-0959
Signe K Skadborg, Simone Maarup, Arianna Draghi, Annie Borch, Sille Hendriksen, Filip Mundt, Vilde Pedersen, Matthias Mann, Ib J Christensen, Jane Skjøth-Ramussen, Christina W Yde, Bjarne W Kristensen, Hans S Poulsen, Benedikte Hasselbalch, Inge M Svane, Ulrik Lassen, Sine R Hadrup

Glioblastoma (GBM) is an aggressive brain tumor with poor prognosis. Although immunotherapy is being explored as a potential treatment option for patients with GBM, it is unclear whether systemic immunotherapy can reach and modify the tumor microenvironment in the brain. We evaluated immune characteristics in patients receiving the anti-PD-1 immune checkpoint inhibitor nivolumab 1 week prior to surgery, compared with control patients receiving salvage resection without prior nivolumab treatment. We observed saturating levels of nivolumab bound to intratumorally and tissue-resident T cells in the brain, implicating saturating levels of nivolumab reaching brain tumors. Following nivolumab treatment, significant changes in T-cell activation and proliferation were observed in the tumor-resident T-cell population, and peripheral T cells upregulated chemokine receptors related to brain homing. A strong nivolumab-driven upregulation in compensatory checkpoint inhibition molecules, i.e., TIGIT, LAG-3, TIM-3, and CTLA-4, was observed, potentially counteracting the treatment effect. Finally, tumor-reactive tumor-infiltrating lymphocytes (TIL) were found in a subset of nivolumab-treated patients with prolonged survival, and neoantigen-reactive T cells were identified in both TILs and blood. This indicates a systemic response toward GBM in a subset of patients, which was further boosted by nivolumab, with T-cell responses toward tumor-derived neoantigens. Our study demonstrates that nivolumab does reach the GBM tumor lesion and enhances antitumor T-cell responses both intratumorally and systemically. However, various anti-inflammatory mechanisms mitigate the clinical efficacy of the anti-PD-1 treatment.

胶质母细胞瘤(GBM)是一种侵袭性脑肿瘤,预后不良。尽管免疫疗法正被探索作为GBM患者的一种潜在治疗选择,但全身性免疫疗法是否能到达并改变脑部肿瘤微环境尚不清楚。我们对手术前一周接受抗 PD1 免疫检查点抑制剂 Nivolumab 治疗的患者的免疫特征进行了评估,并与未接受 Nivolumab 治疗的接受挽救性切除术的对照组患者进行了比较。我们观察到 Nivolumab 与脑内肿瘤内和组织驻留 T 细胞结合的饱和水平,这意味着到达脑肿瘤的 Nivolumab 达到饱和水平。Nivolumab治疗后,在肿瘤常驻T细胞群中观察到T细胞活化和增殖发生了显著变化,外周T细胞上调了与脑归属相关的趋化因子受体。观察到Nivolumab驱动的补偿性检查点抑制分子TIGIT、LAG-3、TIM-3和CTLA-4上调,这可能会抵消治疗效果。最后,在Nivolumab治疗后生存期延长的患者中发现了肿瘤反应性肿瘤浸润淋巴细胞(TILs),并在TILs和血液中发现了新抗原反应性T细胞。这表明在一部分患者中出现了针对GBM的全身反应,Nivolumab进一步增强了这种反应,并出现了针对肿瘤衍生新抗原的T细胞反应。我们的研究表明,Nivolumab 确实能到达 GBM 肿瘤病灶,并增强肿瘤内和全身的抗肿瘤 T 细胞反应。然而,各种抗炎机制削弱了抗 PD1 治疗的临床疗效。
{"title":"Nivolumab Reaches Brain Lesions in Patients with Recurrent Glioblastoma and Induces T-cell Activity and Upregulation of Checkpoint Pathways.","authors":"Signe K Skadborg, Simone Maarup, Arianna Draghi, Annie Borch, Sille Hendriksen, Filip Mundt, Vilde Pedersen, Matthias Mann, Ib J Christensen, Jane Skjøth-Ramussen, Christina W Yde, Bjarne W Kristensen, Hans S Poulsen, Benedikte Hasselbalch, Inge M Svane, Ulrik Lassen, Sine R Hadrup","doi":"10.1158/2326-6066.CIR-23-0959","DOIUrl":"10.1158/2326-6066.CIR-23-0959","url":null,"abstract":"<p><p>Glioblastoma (GBM) is an aggressive brain tumor with poor prognosis. Although immunotherapy is being explored as a potential treatment option for patients with GBM, it is unclear whether systemic immunotherapy can reach and modify the tumor microenvironment in the brain. We evaluated immune characteristics in patients receiving the anti-PD-1 immune checkpoint inhibitor nivolumab 1 week prior to surgery, compared with control patients receiving salvage resection without prior nivolumab treatment. We observed saturating levels of nivolumab bound to intratumorally and tissue-resident T cells in the brain, implicating saturating levels of nivolumab reaching brain tumors. Following nivolumab treatment, significant changes in T-cell activation and proliferation were observed in the tumor-resident T-cell population, and peripheral T cells upregulated chemokine receptors related to brain homing. A strong nivolumab-driven upregulation in compensatory checkpoint inhibition molecules, i.e., TIGIT, LAG-3, TIM-3, and CTLA-4, was observed, potentially counteracting the treatment effect. Finally, tumor-reactive tumor-infiltrating lymphocytes (TIL) were found in a subset of nivolumab-treated patients with prolonged survival, and neoantigen-reactive T cells were identified in both TILs and blood. This indicates a systemic response toward GBM in a subset of patients, which was further boosted by nivolumab, with T-cell responses toward tumor-derived neoantigens. Our study demonstrates that nivolumab does reach the GBM tumor lesion and enhances antitumor T-cell responses both intratumorally and systemically. However, various anti-inflammatory mechanisms mitigate the clinical efficacy of the anti-PD-1 treatment.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11369628/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141417945","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
Iron Boosts Antitumor Type 1 T-cell Responses and Anti-PD1 Immunotherapy. 铁能增强抗肿瘤 1 型 T 细胞反应和抗 PD1 免疫疗法。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-23-0739
Sarah Porte, Alexandra Audemard-Verger, Christian Wu, Aurélie Durand, Théo Level, Léa Giraud, Amélie Lombès, Mathieu Germain, Rémi Pierre, Benjamin Saintpierre, Mireille Lambert, Cédric Auffray, Carole Peyssonnaux, François Goldwasser, Sophie Vaulont, Marie-Clotilde Alves-Guerra, Renaud Dentin, Bruno Lucas, Bruno Martin

Cancers only develop if they escape immunosurveillance, and the success of cancer immunotherapies relies in most cases on their ability to restore effector T-cell functions, particularly IFNγ production. Revolutionizing the treatment of many cancers, immunotherapies targeting immune checkpoints such as PD1 can increase survival and cure patients. Unfortunately, although immunotherapy has greatly improved the prognosis of patients, not all respond to anti-PD1 immunotherapy, making it crucial to identify alternative treatments that could be combined with current immunotherapies to improve their effectiveness. Here, we show that iron supplementation significantly boosts T-cell responses in vivo and in vitro. The boost was associated with a metabolic reprogramming of T cells in favor of lipid oxidation. We also found that the "adjuvant" effect of iron led to a marked slowdown of tumor cell growth after tumor cell line transplantation in mice. Specifically, our results suggest that iron supplementation promotes antitumor responses by increasing IFNγ production by T cells. In addition, iron supplementation improved the efficacy of anti-PD1 cancer immunotherapy in mice. Finally, our study suggests that, in patients with cancer, the quality and efficacy of the antitumor response following anti-PD1 immunotherapy may be modulated by plasma ferritin levels. In summary, our results suggest the benefits of iron supplementation on the reactivation of antitumor responses and support the relevance of a fruitful association between immunotherapy and iron supplementation.

癌症只有在逃避免疫监视的情况下才会发展,而癌症免疫疗法的成功在大多数情况下依赖于其恢复效应T细胞功能的能力,尤其是IFN-γ的产生。以 PD1 等免疫检查点为靶点的免疫疗法为许多癌症的治疗带来了革命性的变化,可以提高患者的生存率并治愈患者。遗憾的是,尽管免疫疗法大大改善了患者的预后,但并非所有患者都对抗抑郁药PD1免疫疗法有反应,因此找出可与现有免疫疗法相结合的替代疗法以提高疗效至关重要。在这里,我们发现补铁能显著提高体内和体外的T细胞反应。这种促进作用与 T 细胞的新陈代谢重编程有关,有利于脂质氧化。我们还发现,铁的 "辅助 "作用导致肿瘤细胞系移植小鼠后肿瘤细胞生长明显减慢。具体来说,我们的研究结果表明,补铁可通过增加 T 细胞产生 IFN-γ 来促进抗肿瘤反应。此外,补铁还能显著提高小鼠抗 PD1 癌症免疫疗法的疗效。最后,我们的研究表明,在癌症患者中,抗 PD1 免疫疗法后抗肿瘤反应的质量和疗效可能受血浆铁蛋白水平的调节。总之,我们的研究结果表明,补铁对重新激活抗肿瘤反应有益,并支持免疫疗法与补铁之间富有成效的关联。
{"title":"Iron Boosts Antitumor Type 1 T-cell Responses and Anti-PD1 Immunotherapy.","authors":"Sarah Porte, Alexandra Audemard-Verger, Christian Wu, Aurélie Durand, Théo Level, Léa Giraud, Amélie Lombès, Mathieu Germain, Rémi Pierre, Benjamin Saintpierre, Mireille Lambert, Cédric Auffray, Carole Peyssonnaux, François Goldwasser, Sophie Vaulont, Marie-Clotilde Alves-Guerra, Renaud Dentin, Bruno Lucas, Bruno Martin","doi":"10.1158/2326-6066.CIR-23-0739","DOIUrl":"10.1158/2326-6066.CIR-23-0739","url":null,"abstract":"<p><p>Cancers only develop if they escape immunosurveillance, and the success of cancer immunotherapies relies in most cases on their ability to restore effector T-cell functions, particularly IFNγ production. Revolutionizing the treatment of many cancers, immunotherapies targeting immune checkpoints such as PD1 can increase survival and cure patients. Unfortunately, although immunotherapy has greatly improved the prognosis of patients, not all respond to anti-PD1 immunotherapy, making it crucial to identify alternative treatments that could be combined with current immunotherapies to improve their effectiveness. Here, we show that iron supplementation significantly boosts T-cell responses in vivo and in vitro. The boost was associated with a metabolic reprogramming of T cells in favor of lipid oxidation. We also found that the \"adjuvant\" effect of iron led to a marked slowdown of tumor cell growth after tumor cell line transplantation in mice. Specifically, our results suggest that iron supplementation promotes antitumor responses by increasing IFNγ production by T cells. In addition, iron supplementation improved the efficacy of anti-PD1 cancer immunotherapy in mice. Finally, our study suggests that, in patients with cancer, the quality and efficacy of the antitumor response following anti-PD1 immunotherapy may be modulated by plasma ferritin levels. In summary, our results suggest the benefits of iron supplementation on the reactivation of antitumor responses and support the relevance of a fruitful association between immunotherapy and iron supplementation.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141442079","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 : 2024-09-03 DOI: 10.1158/2326-6066.CIR-12-9-WWR
{"title":"A Sampling of Highlights from the Literature.","authors":"","doi":"10.1158/2326-6066.CIR-12-9-WWR","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-12-9-WWR","url":null,"abstract":"","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142119066","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
Knocking Out CD70 Rescues CD70-Specific NanoCAR T Cells from Antigen-Induced Exhaustion. 敲除 CD70 可使 CD70 特异性 nanoCAR T 细胞从抗原诱导的衰竭中解救出来。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-23-0677
Stijn De Munter, Juliane L Buhl, Laurenz De Cock, Alexander Van Parys, Willem Daneels, Eva Pascal, Lucas Deseins, Joline Ingels, Glenn Goetgeluk, Hanne Jansen, Lore Billiet, Melissa Pille, Julie Van Duyse, Sarah Bonte, Niels Vandamme, Jo Van Dorpe, Fritz Offner, Georges Leclercq, Tom Taghon, Erik Depla, Jan Tavernier, Tessa Kerre, Jarno Drost, Bart Vandekerckhove

CD70 is an attractive target for chimeric antigen receptor (CAR) T-cell therapy for the treatment of both solid and liquid malignancies. However, the functionality of CD70-specific CAR T cells is modest. We optimized a CD70-specific VHH-based CAR (nanoCAR). We evaluated the nanoCARs in clinically relevant models in vitro, using co-cultures of CD70-specific nanoCAR T cells with malignant rhabdoid tumor organoids, and in vivo, using a diffuse large B-cell lymphoma patient-derived xenograft (PDX) model. Although the nanoCAR T cells were highly efficient in organoid co-cultures, they showed only modest efficacy in the PDX model. We determined that fratricide was not causing this loss in efficacy but rather CD70 interaction in cis with the nanoCAR-induced exhaustion. Knocking out CD70 in nanoCAR T cells using CRISPR/Cas9 resulted in dramatically enhanced functionality in the diffuse large B-cell lymphoma PDX model. Through single-cell transcriptomics, we obtained evidence that CD70 knockout CD70-specific nanoCAR T cells were protected from antigen-induced exhaustion. In addition, we demonstrated that wild-type CD70-specific nanoCAR T cells already exhibited signs of exhaustion shortly after production. Their gene signature strongly overlapped with gene signatures of exhausted CAR T cells. Conversely, the gene signature of knockout CD70-specific nanoCAR T cells overlapped with the gene signature of CAR T-cell infusion products leading to complete responses in chronic lymphatic leukemia patients. Our data show that CARs targeting endogenous T-cell antigens negatively affect CAR T-cell functionality by inducing an exhausted state, which can be overcome by knocking out the specific target.

对于治疗实体和液体恶性肿瘤的嵌合抗原受体(CAR)T细胞疗法来说,CD70是一个极具吸引力的靶点。然而,CD70 特异性 CAR T 细胞的功能并不强大。我们优化了一种基于 VHH 的 CD70 特异性 CAR(nanoCAR)。我们在体外使用 CD70 特异性 nanoCAR T 细胞与恶性横纹肌瘤器官组织共培养,在体内使用弥漫大 B 细胞淋巴瘤(DLBCL)患者衍生异种移植(PDX)模型,在临床相关模型中对 nanoCAR 进行了评估。虽然纳米CAR T细胞在类器官共培养中效率很高,但在PDX模型中仅表现出适度的疗效。我们确定,"自相残杀 "并不是导致疗效下降的原因,而是 CD70 与 nanoCAR 的顺式相互作用导致了细胞衰竭。使用 CRISPR/Cas9 基因敲除 nanoCAR T 细胞中的 CD70 后,DLBCL PDX 模型的功能显著增强。通过单细胞转录组学,我们获得了CD70敲除(KO)的CD70特异性纳米CAR T细胞免受抗原诱导的衰竭的证据。此外,我们还证明,WT CD70特异性纳米CAR T细胞在产生后不久就出现了衰竭迹象。它们的基因特征与衰竭的 CAR T 细胞的基因特征高度重叠。另一方面,KO CD70 特异性 nanoCAR T 细胞的基因特征与导致慢性淋巴白血病患者完全应答的 CAR T 细胞输注产物的基因特征重叠。我们的数据表明,以内源性 T 细胞抗原为靶点的 CAR 通过诱导衰竭状态对 CAR T 细胞的功能产生负面影响,而通过敲除特异性靶点可以克服这种影响。
{"title":"Knocking Out CD70 Rescues CD70-Specific NanoCAR T Cells from Antigen-Induced Exhaustion.","authors":"Stijn De Munter, Juliane L Buhl, Laurenz De Cock, Alexander Van Parys, Willem Daneels, Eva Pascal, Lucas Deseins, Joline Ingels, Glenn Goetgeluk, Hanne Jansen, Lore Billiet, Melissa Pille, Julie Van Duyse, Sarah Bonte, Niels Vandamme, Jo Van Dorpe, Fritz Offner, Georges Leclercq, Tom Taghon, Erik Depla, Jan Tavernier, Tessa Kerre, Jarno Drost, Bart Vandekerckhove","doi":"10.1158/2326-6066.CIR-23-0677","DOIUrl":"10.1158/2326-6066.CIR-23-0677","url":null,"abstract":"<p><p>CD70 is an attractive target for chimeric antigen receptor (CAR) T-cell therapy for the treatment of both solid and liquid malignancies. However, the functionality of CD70-specific CAR T cells is modest. We optimized a CD70-specific VHH-based CAR (nanoCAR). We evaluated the nanoCARs in clinically relevant models in vitro, using co-cultures of CD70-specific nanoCAR T cells with malignant rhabdoid tumor organoids, and in vivo, using a diffuse large B-cell lymphoma patient-derived xenograft (PDX) model. Although the nanoCAR T cells were highly efficient in organoid co-cultures, they showed only modest efficacy in the PDX model. We determined that fratricide was not causing this loss in efficacy but rather CD70 interaction in cis with the nanoCAR-induced exhaustion. Knocking out CD70 in nanoCAR T cells using CRISPR/Cas9 resulted in dramatically enhanced functionality in the diffuse large B-cell lymphoma PDX model. Through single-cell transcriptomics, we obtained evidence that CD70 knockout CD70-specific nanoCAR T cells were protected from antigen-induced exhaustion. In addition, we demonstrated that wild-type CD70-specific nanoCAR T cells already exhibited signs of exhaustion shortly after production. Their gene signature strongly overlapped with gene signatures of exhausted CAR T cells. Conversely, the gene signature of knockout CD70-specific nanoCAR T cells overlapped with the gene signature of CAR T-cell infusion products leading to complete responses in chronic lymphatic leukemia patients. Our data show that CARs targeting endogenous T-cell antigens negatively affect CAR T-cell functionality by inducing an exhausted state, which can be overcome by knocking out the specific target.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141316752","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
CAR T cells engineered to secrete IFN-κ induce tumor ferroptosis via an IFNAR/STAT1/ACSL4 axis. 经改造可分泌 IFN-κ 的 CAR T 细胞通过 IFNAR/STAT1/ACSL4 轴诱导肿瘤铁变态反应。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-26 DOI: 10.1158/2326-6066.CIR-24-0130
Yaoxin Gao, Shasha Liu, Yifan Huang, Hui Wang, Yuyu Zhao, Xuyang Cui, Yajing Peng, Feng Li, Yi Zhang

Ferroptosis is an iron-dependent form of cell death that influences cancer immunity. Therapeutic modulation of ferroptosis is considered a potential strategy to enhance the efficacy of other cancer therapies, including immunotherapies such as chimeric antigen receptor (CAR) T cell therapy. In this study, we demonstrated that IFN-κ influenced the induction of ferroptosis. IFN-κ could enhance the sensitivity of tumor cells to ferroptosis induced by the small molecule compound erastin and the polyunsaturated fatty acid arachidonic acid. Mechanistically, IFN-κ in combination with arachidonic acid induced immunogenic tumor ferroptosis via an IFNAR/STAT1/ACSL4 axis. Moreover, CAR T cells engineered to express IFN-κ showed increased antitumor efficiency against H460 cells (antigen positive) and H322 cells (antigen negative) both in vitro and in vivo. We conclude that IFN-κ is a potential cytokine that could be harnessed to enhance the antitumor function of CAR T cells by inducing tumor ferroptosis.

铁突变是一种影响癌症免疫的铁依赖性细胞死亡形式。对铁凋亡的治疗调节被认为是提高其他癌症疗法疗效的一种潜在策略,包括嵌合抗原受体(CAR)T细胞疗法等免疫疗法。在这项研究中,我们证明了 IFN-κ 对铁卟啉诱导的影响。IFN-κ能增强肿瘤细胞对小分子化合物麦拉宁和多不饱和脂肪酸花生四烯酸诱导的铁变态反应的敏感性。从机理上讲,IFN-κ与花生四烯酸结合可通过IFNAR/STAT1/ACSL4轴诱导免疫原性肿瘤铁中毒。此外,表达 IFN-κ 的 CAR T 细胞在体外和体内对 H460 细胞(抗原阳性)和 H322 细胞(抗原阴性)的抗肿瘤效率都有所提高。我们的结论是,IFN-κ是一种潜在的细胞因子,可以通过诱导肿瘤铁变态反应来增强CAR T细胞的抗肿瘤功能。
{"title":"CAR T cells engineered to secrete IFN-κ induce tumor ferroptosis via an IFNAR/STAT1/ACSL4 axis.","authors":"Yaoxin Gao, Shasha Liu, Yifan Huang, Hui Wang, Yuyu Zhao, Xuyang Cui, Yajing Peng, Feng Li, Yi Zhang","doi":"10.1158/2326-6066.CIR-24-0130","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-24-0130","url":null,"abstract":"<p><p>Ferroptosis is an iron-dependent form of cell death that influences cancer immunity. Therapeutic modulation of ferroptosis is considered a potential strategy to enhance the efficacy of other cancer therapies, including immunotherapies such as chimeric antigen receptor (CAR) T cell therapy. In this study, we demonstrated that IFN-κ influenced the induction of ferroptosis. IFN-κ could enhance the sensitivity of tumor cells to ferroptosis induced by the small molecule compound erastin and the polyunsaturated fatty acid arachidonic acid. Mechanistically, IFN-κ in combination with arachidonic acid induced immunogenic tumor ferroptosis via an IFNAR/STAT1/ACSL4 axis. Moreover, CAR T cells engineered to express IFN-κ showed increased antitumor efficiency against H460 cells (antigen positive) and H322 cells (antigen negative) both in vitro and in vivo. We conclude that IFN-κ is a potential cytokine that could be harnessed to enhance the antitumor function of CAR T cells by inducing tumor ferroptosis.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142072107","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
Age-associated contraction of tumor-specific T cells impairs antitumor immunity. 与年龄相关的肿瘤特异性 T 细胞收缩会损害抗肿瘤免疫力。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-24 DOI: 10.1158/2326-6066.CIR-24-0463
Peter Georgiev, SeongJun Han, Amy Y Huang, Thao H Nguyen, Jefte M Drijvers, Hannah Creasey, Joseph A Pereira, Cong-Hui Yao, Joon Seok Park, Thomas S Conway, Megan E Fung, Dan Liang, Michael Peluso, Shakchhi Joshi, Jared H Rowe, Brian C Miller, Gordon J Freeman, Arlene H Sharpe, Marcia C Haigis, Alison E Ringel

Progressive decline of the adaptive immune system with increasing age coincides with a sharp increase in cancer incidence. In this study, we set out to understand whether deficits in antitumor immunity with advanced age promote tumor progression and/or drive resistance to immunotherapy. We found that multiple syngeneic cancers grew more rapidly in aged versus young adult mice, driven by dysfunctional CD8+ T-cell responses. By systematically mapping immune cell profiles within tumors, we identified loss of tumor antigen-specific CD8+ T cells as a primary feature accelerating the growth of tumors in aged mice and driving resistance to immunotherapy. When antigen-specific T cells from young adult mice were administered to aged mice, tumor outgrowth was delayed and the aged animals became sensitive to PD-1 blockade. These studies reveal how age-associated CD8+ T-cell dysfunction may license tumorigenesis in elderly patients and have important implications for the use of aged mice as pre-clinical models of aging and cancer.

随着年龄的增长,适应性免疫系统逐渐衰退,同时癌症发病率也急剧上升。在这项研究中,我们试图了解随着年龄的增长,抗肿瘤免疫系统的缺陷是否会促进肿瘤的发展和/或驱动对免疫疗法的抵抗。我们发现,在 CD8+ T 细胞反应失调的驱动下,老年小鼠与年轻成年小鼠相比,多种共生癌症的生长速度更快。通过系统地绘制肿瘤内的免疫细胞图谱,我们发现肿瘤抗原特异性 CD8+ T 细胞的缺失是加速老年小鼠肿瘤生长和驱动免疫疗法抗药性的主要特征。当将来自年轻成年小鼠的抗原特异性 T 细胞注射给老年小鼠时,肿瘤的生长被延缓,而且老年动物对 PD-1 阻断剂变得敏感。这些研究揭示了与年龄相关的 CD8+ T 细胞功能障碍如何许可老年患者的肿瘤发生,并对使用老年小鼠作为衰老和癌症的临床前模型具有重要意义。
{"title":"Age-associated contraction of tumor-specific T cells impairs antitumor immunity.","authors":"Peter Georgiev, SeongJun Han, Amy Y Huang, Thao H Nguyen, Jefte M Drijvers, Hannah Creasey, Joseph A Pereira, Cong-Hui Yao, Joon Seok Park, Thomas S Conway, Megan E Fung, Dan Liang, Michael Peluso, Shakchhi Joshi, Jared H Rowe, Brian C Miller, Gordon J Freeman, Arlene H Sharpe, Marcia C Haigis, Alison E Ringel","doi":"10.1158/2326-6066.CIR-24-0463","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-24-0463","url":null,"abstract":"<p><p>Progressive decline of the adaptive immune system with increasing age coincides with a sharp increase in cancer incidence. In this study, we set out to understand whether deficits in antitumor immunity with advanced age promote tumor progression and/or drive resistance to immunotherapy. We found that multiple syngeneic cancers grew more rapidly in aged versus young adult mice, driven by dysfunctional CD8+ T-cell responses. By systematically mapping immune cell profiles within tumors, we identified loss of tumor antigen-specific CD8+ T cells as a primary feature accelerating the growth of tumors in aged mice and driving resistance to immunotherapy. When antigen-specific T cells from young adult mice were administered to aged mice, tumor outgrowth was delayed and the aged animals became sensitive to PD-1 blockade. These studies reveal how age-associated CD8+ T-cell dysfunction may license tumorigenesis in elderly patients and have important implications for the use of aged mice as pre-clinical models of aging and cancer.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142072106","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
CD36+ pro-inflammatory macrophages interact with ZCCHC12+ tumor cells in papillary thyroid cancer promoting tumor progression and recurrence. CD36+ 促炎巨噬细胞与甲状腺乳头状癌中的 ZCCHC12+ 肿瘤细胞相互作用,促进肿瘤进展和复发。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-23 DOI: 10.1158/2326-6066.CIR-23-1047
Xin Zhang, Limei Guo, Wenyu Tian, Ying Yang, Yue Yin, Yaruo Qiu, Weixuan Wang, Yang Li, Guangze Zhang, Xuyang Zhao, Guangxi Wang, Zhiqiang Lin, Meng Yang, Wei Zhao, Dan Lu

Local recurrence and distal metastasis negatively impact the survival and quality of life in patients with papillary thyroid cancer (PTC). Therefore, identifying potential biomarkers and therapeutic targets for PTC is clinically crucial. In this study, we performed a multi-omics analysis that identified a subset of CD36+ pro-inflammatory macrophages within the tumor microenvironment of PTC. The recruitment of CD36+ macrophages to pre-malignant regions strongly correlated with unfavorable outcomes in PTC and the presence of tumor-infiltrating CD36+ macrophages was determined to be a risk factor for recurrence. The CD36+ macrophages exhibited interactions with metabolically active ZCCHC12+ tumor cells. By secreting SPP1, the CD36+ macrophages activated the PI3K-AKT signaling pathway, thereby promoting proliferation of the cancer cells. Dysregulation of iodine metabolism was closely related to the acquisition of the pro-inflammatory phenotype in macrophages. Iodine supplementation inhibited the activation of pro-inflammatory signaling and impeded the development of CD36+ macrophages by enhancing DUSP2 expression. Overall, our findings shed light on the intricate crosstalk between CD36+ macrophages and ZCCHC12+ tumor cells, providing valuable insights for the treatment and prognosis of PTC.

局部复发和远处转移会对甲状腺乳头状癌(PTC)患者的生存和生活质量产生负面影响。因此,确定 PTC 的潜在生物标志物和治疗靶点在临床上至关重要。在这项研究中,我们进行了一项多组学分析,发现了PTC肿瘤微环境中的CD36+促炎巨噬细胞亚群。CD36+巨噬细胞被招募到恶性肿瘤前区域与PTC的不良预后密切相关,肿瘤浸润CD36+巨噬细胞的存在被确定为复发的风险因素。CD36+ 巨噬细胞与代谢活跃的 ZCCHC12+ 肿瘤细胞相互作用。通过分泌 SPP1,CD36+ 巨噬细胞激活了 PI3K-AKT 信号通路,从而促进了癌细胞的增殖。碘代谢失调与巨噬细胞获得促炎表型密切相关。补碘可抑制促炎信号的激活,并通过增强 DUSP2 的表达阻碍 CD36+ 巨噬细胞的发展。总之,我们的研究结果揭示了 CD36+ 巨噬细胞和 ZCCHC12+ 肿瘤细胞之间错综复杂的相互影响,为 PTC 的治疗和预后提供了有价值的见解。
{"title":"CD36+ pro-inflammatory macrophages interact with ZCCHC12+ tumor cells in papillary thyroid cancer promoting tumor progression and recurrence.","authors":"Xin Zhang, Limei Guo, Wenyu Tian, Ying Yang, Yue Yin, Yaruo Qiu, Weixuan Wang, Yang Li, Guangze Zhang, Xuyang Zhao, Guangxi Wang, Zhiqiang Lin, Meng Yang, Wei Zhao, Dan Lu","doi":"10.1158/2326-6066.CIR-23-1047","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-23-1047","url":null,"abstract":"<p><p>Local recurrence and distal metastasis negatively impact the survival and quality of life in patients with papillary thyroid cancer (PTC). Therefore, identifying potential biomarkers and therapeutic targets for PTC is clinically crucial. In this study, we performed a multi-omics analysis that identified a subset of CD36+ pro-inflammatory macrophages within the tumor microenvironment of PTC. The recruitment of CD36+ macrophages to pre-malignant regions strongly correlated with unfavorable outcomes in PTC and the presence of tumor-infiltrating CD36+ macrophages was determined to be a risk factor for recurrence. The CD36+ macrophages exhibited interactions with metabolically active ZCCHC12+ tumor cells. By secreting SPP1, the CD36+ macrophages activated the PI3K-AKT signaling pathway, thereby promoting proliferation of the cancer cells. Dysregulation of iodine metabolism was closely related to the acquisition of the pro-inflammatory phenotype in macrophages. Iodine supplementation inhibited the activation of pro-inflammatory signaling and impeded the development of CD36+ macrophages by enhancing DUSP2 expression. Overall, our findings shed light on the intricate crosstalk between CD36+ macrophages and ZCCHC12+ tumor cells, providing valuable insights for the treatment and prognosis of PTC.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142043866","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 single-cell analysis of the NK-cell landscape reveals that dietary restriction boosts NK cell antitumor immunity via Eomesdermin. 对 NK 细胞格局的单细胞分析表明,饮食限制可通过 Eomesdermin 增强 NK 细胞的抗肿瘤免疫力。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-16 DOI: 10.1158/2326-6066.CIR-23-0944
Junming He, Donglin Chen, Wei Xiong, Yuande Wang, Shasha Chen, Meixiang Yang, Zhongjun Dong

Abnormal metabolism in tumor cells represents a potential target for tumor therapy. In this regard, dietary restriction (DR) or its combination with anticancer drugs is of interest as it can impede the growth of tumor cells. In addition to its effects on tumor cell, DR also plays an extrinsic role in restricting tumor growth by regulating immune cells. Natural killer (NK) cells are innate immune cells involved in tumor immunosurveillance. However, it remains uncertain whether DR can assist NK cells in controlling tumor growth. Herein, we demonstrate that DR effectively inhibits metastasis of melanoma cells to the lung. Consistent with this, the regression of tumors induced by DR was minimal in mice lacking NK cells. Single-cell RNA sequencing analysis revealed that DR enriched a rejuvenated subset of CD27+CD11b+ NK cells. Mechanistically, DR activated a regulatory network involving the transcription factor Eomesodermin (Eomes), which is essential for NK-cell development. Firstly, DR promoted the expression of Eomes by optimizing mTORC1 signaling. The upregulation of Eomes revived the subset of functional CD27+CD11b+ NK cells by counteracting the expression of T-bet and downstream Zeb2. Moreover, DR enhanced the function and chemotaxis of NK cells by increasing the accessibility of Eomes to chromatin, leading to elevated expression of adhesion molecules and chemokines. Consequently, we conclude that DR therapy enhances tumor immunity through non-tumor autonomous mechanisms, including promoting NK-cell tumor immunosurveillance and activation.

肿瘤细胞代谢异常是肿瘤治疗的潜在靶点。在这方面,饮食限制(DR)或其与抗癌药物的结合能够阻碍肿瘤细胞的生长,因此备受关注。除了对肿瘤细胞的影响外,DR 还通过调节免疫细胞在限制肿瘤生长方面发挥外在作用。自然杀伤(NK)细胞是参与肿瘤免疫监视的先天性免疫细胞。然而,DR 是否能帮助 NK 细胞控制肿瘤生长仍不确定。在此,我们证明 DR 能有效抑制黑色素瘤细胞向肺部转移。与此相一致的是,在缺乏 NK 细胞的小鼠体内,DR 诱导的肿瘤消退效果甚微。单细胞 RNA 测序分析表明,DR 富集了 CD27+CD11b+ NK 细胞的年轻化亚群。从机理上讲,DR激活了涉及转录因子Eomesodermin(Eomes)的调控网络,而Eomes对NK细胞的发育至关重要。首先,DR通过优化mTORC1信号来促进Eomes的表达。此外,DR通过增加Eomes对染色质的可及性,导致粘附分子和趋化因子的表达增加,从而增强了NK细胞的功能和趋化性。因此,我们得出结论:DR疗法通过非肿瘤自主机制增强肿瘤免疫力,包括促进NK细胞的肿瘤免疫监视和激活。
{"title":"A single-cell analysis of the NK-cell landscape reveals that dietary restriction boosts NK cell antitumor immunity via Eomesdermin.","authors":"Junming He, Donglin Chen, Wei Xiong, Yuande Wang, Shasha Chen, Meixiang Yang, Zhongjun Dong","doi":"10.1158/2326-6066.CIR-23-0944","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-23-0944","url":null,"abstract":"<p><p>Abnormal metabolism in tumor cells represents a potential target for tumor therapy. In this regard, dietary restriction (DR) or its combination with anticancer drugs is of interest as it can impede the growth of tumor cells. In addition to its effects on tumor cell, DR also plays an extrinsic role in restricting tumor growth by regulating immune cells. Natural killer (NK) cells are innate immune cells involved in tumor immunosurveillance. However, it remains uncertain whether DR can assist NK cells in controlling tumor growth. Herein, we demonstrate that DR effectively inhibits metastasis of melanoma cells to the lung. Consistent with this, the regression of tumors induced by DR was minimal in mice lacking NK cells. Single-cell RNA sequencing analysis revealed that DR enriched a rejuvenated subset of CD27+CD11b+ NK cells. Mechanistically, DR activated a regulatory network involving the transcription factor Eomesodermin (Eomes), which is essential for NK-cell development. Firstly, DR promoted the expression of Eomes by optimizing mTORC1 signaling. The upregulation of Eomes revived the subset of functional CD27+CD11b+ NK cells by counteracting the expression of T-bet and downstream Zeb2. Moreover, DR enhanced the function and chemotaxis of NK cells by increasing the accessibility of Eomes to chromatin, leading to elevated expression of adhesion molecules and chemokines. Consequently, we conclude that DR therapy enhances tumor immunity through non-tumor autonomous mechanisms, including promoting NK-cell tumor immunosurveillance and activation.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141987431","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