首页 > 最新文献

Cancer immunology research最新文献

英文 中文
Immunotherapy Inhibits Tumor Cholesterol Synthesis via the IFN-γ-ΙRF1-SREBF2 Axis. 免疫疗法通过IFN-γ-ΙRF1-SREBF2轴抑制肿瘤胆固醇合成。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-09 DOI: 10.1158/2326-6066.CIR-25-0242
Liping Xu, Xiaomin Zhang, Xiaowei Lai, Hongyuan Chen, Shuangye Liao, Mingzeng Chang, Xiaoqing Wu, Wen Rui, Juan Yang, Danyang Wang, Chengqi Gao, Ziqian Fang, Jianeng Zhang, Wende Li, Bo Li, Xiaojun Xia, Penghui Zhou

Tumor cells employ metabolic mechanisms to limit antitumor immunity and promote resistance to immunotherapy. However, how immunotherapy modulates tumor metabolism remains unclear. Here, we demonstrated that anti-PD-1 treatment regulated cholesterol biosynthesis in cancer cells through the effector cytokine interferon IFN-γ. Mechanistically, IFN-γ-induced IRF1 transcriptionally suppresses the expression of SREBF2, a master regulator of cholesterol synthesis. Reduced cholesterol content inhibited tumor growth and sensitized tumor cells to statins, drugs lowering cholesterol. Overall, our study reveals that IFN-γ-mediated inhibition of cholesterol biosynthesis in tumor cells is an important antitumor mechanism of immunotherapy.

肿瘤细胞利用代谢机制限制抗肿瘤免疫并促进对免疫治疗的抵抗。然而,免疫疗法如何调节肿瘤代谢仍不清楚。在这里,我们证明了抗pd -1治疗通过效应细胞因子干扰素IFN-γ调节癌细胞中的胆固醇生物合成。从机制上讲,IFN-γ诱导的IRF1转录抑制SREBF2的表达,SREBF2是胆固醇合成的主要调节因子。降低胆固醇含量抑制肿瘤生长,使肿瘤细胞对他汀类降胆固醇药物敏感。综上所述,我们的研究揭示了IFN-γ介导的抑制肿瘤细胞胆固醇生物合成是免疫治疗的重要抗肿瘤机制。
{"title":"Immunotherapy Inhibits Tumor Cholesterol Synthesis via the IFN-γ-ΙRF1-SREBF2 Axis.","authors":"Liping Xu, Xiaomin Zhang, Xiaowei Lai, Hongyuan Chen, Shuangye Liao, Mingzeng Chang, Xiaoqing Wu, Wen Rui, Juan Yang, Danyang Wang, Chengqi Gao, Ziqian Fang, Jianeng Zhang, Wende Li, Bo Li, Xiaojun Xia, Penghui Zhou","doi":"10.1158/2326-6066.CIR-25-0242","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-25-0242","url":null,"abstract":"<p><p>Tumor cells employ metabolic mechanisms to limit antitumor immunity and promote resistance to immunotherapy. However, how immunotherapy modulates tumor metabolism remains unclear. Here, we demonstrated that anti-PD-1 treatment regulated cholesterol biosynthesis in cancer cells through the effector cytokine interferon IFN-γ. Mechanistically, IFN-γ-induced IRF1 transcriptionally suppresses the expression of SREBF2, a master regulator of cholesterol synthesis. Reduced cholesterol content inhibited tumor growth and sensitized tumor cells to statins, drugs lowering cholesterol. Overall, our study reveals that IFN-γ-mediated inhibition of cholesterol biosynthesis in tumor cells is an important antitumor mechanism of immunotherapy.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145707461","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 universal boosting strategy for adoptive T cell therapy using a paired vaccine/chimeric antigen receptor. 使用配对疫苗/嵌合抗原受体的过继性T细胞治疗的通用增强策略。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-08 DOI: 10.1158/2326-6066.CIR-25-0070
Rebecca Burchett, Claire G Morris, Mira Ishak, Nickolas J Serniuck, Derek T Cummings, Christopher L Baker, Ricardo Marius, Natasha Kazdhan, Christopher M Silvestri, John Bell, Brian D Lichty, Scott R Walsh, Yonghong Wan, Joanne A Hammill, Jonathan L Bramson

Vaccines that encode tumour-associated antigens are potent boosting agents for adoptively transferred tumour-specific T cells. Employing vaccines to boost adoptively transferred tumour-reactive T cells relies on a priori knowledge of tumour epitopes, isolation of matched epitope-specific T cells, and personalized vaccines, all of which limit clinical feasibility. Here, we investigated a universal strategy for boosting transferred tumour-specific T cells where boosting is provided through a chimeric antigen receptor (CAR) that is paired with a vaccine encoding the CAR target antigen. To this end, we developed and employed a model wherein murine T cells expressing a TCR specific for antigen on syngeneic tumours were engineered with boosting CARs against a distinct surrogate boosting antigen for studies in immunocompetent hosts. Boosting CAR-engineered tumour-specific T cells with paired vesicular stomatitis virus (VSV) vaccines was associated with robust T cell expansion and delayed tumour progression in the absence of prior lymphodepletion. CAR-T cell expansion and antitumour function was further enhanced by blocking IFNAR1. However, vaccine-boosted CAR-T cells rapidly contracted and antigen-positive tumours re-emerged. In contrast, when the same T cells were boosted with a vaccine encoding antigen that stimulates through the TCR, the adoptively transferred T cells displayed improved persistence, tumour-specific endogenous cells expanded in parallel, and tumour cells carrying the antigen target were completely eradicated. Our findings underscore the need for further research into CAR-mediated vaccine boosting, how this differs mechanistically from TCR-mediated boosting, and the importance of engaging endogenous tumour-reactive T cells during vaccination to achieve long-term tumour control.

编码肿瘤相关抗原的疫苗是过继性转移肿瘤特异性T细胞的有效促进剂。使用疫苗来促进过继转移的肿瘤反应性T细胞依赖于对肿瘤表位的先验知识、匹配表位特异性T细胞的分离和个性化疫苗,所有这些都限制了临床可行性。在这里,我们研究了一种增强转移肿瘤特异性T细胞的通用策略,其中通过与编码CAR靶抗原的疫苗配对的嵌合抗原受体(CAR)提供增强。为此,我们开发并采用了一种模型,在该模型中,表达同源肿瘤抗原特异性TCR的小鼠T细胞被改造成增强car,以对抗不同的替代增强抗原,用于免疫活性宿主的研究。用配对的水疱性口炎病毒(VSV)疫苗增强car工程肿瘤特异性T细胞,在没有事先淋巴细胞清除的情况下,与强大的T细胞扩增和延迟肿瘤进展相关。阻断IFNAR1可进一步增强CAR-T细胞扩增和抗肿瘤功能。然而,疫苗增强的CAR-T细胞迅速收缩,抗原阳性肿瘤再次出现。相比之下,当同样的T细胞被编码抗原的疫苗增强时,通过TCR刺激,过继转移的T细胞表现出更好的持久性,肿瘤特异性内源性细胞平行扩增,携带抗原靶标的肿瘤细胞被完全根除。我们的研究结果强调需要进一步研究car介导的疫苗增强,这与tcr介导的增强在机制上有何不同,以及在疫苗接种期间参与内源性肿瘤反应性T细胞以实现长期肿瘤控制的重要性。
{"title":"A universal boosting strategy for adoptive T cell therapy using a paired vaccine/chimeric antigen receptor.","authors":"Rebecca Burchett, Claire G Morris, Mira Ishak, Nickolas J Serniuck, Derek T Cummings, Christopher L Baker, Ricardo Marius, Natasha Kazdhan, Christopher M Silvestri, John Bell, Brian D Lichty, Scott R Walsh, Yonghong Wan, Joanne A Hammill, Jonathan L Bramson","doi":"10.1158/2326-6066.CIR-25-0070","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-25-0070","url":null,"abstract":"<p><p>Vaccines that encode tumour-associated antigens are potent boosting agents for adoptively transferred tumour-specific T cells. Employing vaccines to boost adoptively transferred tumour-reactive T cells relies on a priori knowledge of tumour epitopes, isolation of matched epitope-specific T cells, and personalized vaccines, all of which limit clinical feasibility. Here, we investigated a universal strategy for boosting transferred tumour-specific T cells where boosting is provided through a chimeric antigen receptor (CAR) that is paired with a vaccine encoding the CAR target antigen. To this end, we developed and employed a model wherein murine T cells expressing a TCR specific for antigen on syngeneic tumours were engineered with boosting CARs against a distinct surrogate boosting antigen for studies in immunocompetent hosts. Boosting CAR-engineered tumour-specific T cells with paired vesicular stomatitis virus (VSV) vaccines was associated with robust T cell expansion and delayed tumour progression in the absence of prior lymphodepletion. CAR-T cell expansion and antitumour function was further enhanced by blocking IFNAR1. However, vaccine-boosted CAR-T cells rapidly contracted and antigen-positive tumours re-emerged. In contrast, when the same T cells were boosted with a vaccine encoding antigen that stimulates through the TCR, the adoptively transferred T cells displayed improved persistence, tumour-specific endogenous cells expanded in parallel, and tumour cells carrying the antigen target were completely eradicated. Our findings underscore the need for further research into CAR-mediated vaccine boosting, how this differs mechanistically from TCR-mediated boosting, and the importance of engaging endogenous tumour-reactive T cells during vaccination to achieve long-term tumour control.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145699722","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
T cell priming by high avidity neoantigens in lymph nodes augments adoptive immunotherapy. 淋巴结中高亲和力新抗原的T细胞启动增强了过继免疫治疗。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-05 DOI: 10.1158/2326-6066.CIR-25-0514
Megen C Wittling, Amalia M Rivera Reyes, Megan M Wyatt, Anna C Cole, Aubrey S Smith, Guillermo O Rangel Rivera, James H Carmouche, Kailey G Diatikar, Ayana T Ruffin, Michael B Ware, Frances J Bennett, Connor J Dwyer, Riley M F Pihl, Soundharya Kumaresan, Gregory B Lesinski, Chrystal M Paulos, Hannah M Knochelmann

Adoptive transfer of T lymphocytes specific for tumor-associated neoantigens can elicit immunity against solid tumors in patients. However, how these antigens impact T cell function, effector differentiation, and persistence remains unclear. We examined how an identical CD8+ T cell product was shaped by melanoma expressing either a low-avidity tumor-associated antigen or high-avidity neoantigen, and kinetically profiled T cell differentiation in these two contexts across host tissues. High-avidity neoantigen expression was sufficient to activate naïve CD8+ T cells - leading to robust tumor regression and long-term protective immunity upon tumor rechallenge. Mechanistically, transferred naïve CD8+ T cells reacting to high-avidity neoantigen exhibited enhanced cytokine production, heightened effector function, and sustained persistence compared to the low-avidity wild-type tumors. Antitumor activity to these high-avidity tumors was preserved even in the absence of functional host T and B lymphocytes, and early lymph node trafficking was found to be essential for ACT efficacy. Expanded effector or stem-memory T cells were compared to the naïve pmel-1 T cell product. Stem-memory but not effector-memory cells exhibited similar antitumor efficacy and lymph node trafficking patterns to the naïve cells in mice with high-avidity neoantigen tumors. These findings highlight how differential tumor antigens shape divergent cellular fate and uncover a critical role of T cell trafficking in lymph nodes in shaping high-avidity neoantigen-specific antitumor responses.

肿瘤相关新抗原特异性T淋巴细胞的过继转移可引起患者对实体瘤的免疫。然而,这些抗原如何影响T细胞功能、效应分化和持久性仍不清楚。我们研究了相同的CD8+ T细胞产物是如何通过表达低亲和力肿瘤相关抗原或高亲和力新抗原的黑色素瘤形成的,并在这两种情况下在宿主组织中动态地描述了T细胞分化。高频率的新抗原表达足以激活naïve CD8+ T细胞,从而在肿瘤再攻击时实现强劲的肿瘤消退和长期的保护性免疫。在机制上,与低亲和力的野生型肿瘤相比,转移的naïve CD8+ T细胞对高亲和力的新抗原反应,表现出增强的细胞因子产生,增强的效应功能和持续的持久性。即使在缺乏功能性宿主T淋巴细胞和B淋巴细胞的情况下,对这些高发性肿瘤的抗肿瘤活性仍然保持不变,并且发现早期淋巴结运输对于ACT的疗效至关重要。将扩增效应T细胞或干细胞记忆T细胞与naïve pmel-1 T细胞产物进行比较。干细胞记忆而非效应记忆细胞表现出与高亲和性新抗原肿瘤小鼠naïve细胞相似的抗肿瘤功效和淋巴结运输模式。这些发现强调了不同的肿瘤抗原如何塑造不同的细胞命运,并揭示了淋巴结中T细胞运输在形成高亲和力的新抗原特异性抗肿瘤反应中的关键作用。
{"title":"T cell priming by high avidity neoantigens in lymph nodes augments adoptive immunotherapy.","authors":"Megen C Wittling, Amalia M Rivera Reyes, Megan M Wyatt, Anna C Cole, Aubrey S Smith, Guillermo O Rangel Rivera, James H Carmouche, Kailey G Diatikar, Ayana T Ruffin, Michael B Ware, Frances J Bennett, Connor J Dwyer, Riley M F Pihl, Soundharya Kumaresan, Gregory B Lesinski, Chrystal M Paulos, Hannah M Knochelmann","doi":"10.1158/2326-6066.CIR-25-0514","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-25-0514","url":null,"abstract":"<p><p>Adoptive transfer of T lymphocytes specific for tumor-associated neoantigens can elicit immunity against solid tumors in patients. However, how these antigens impact T cell function, effector differentiation, and persistence remains unclear. We examined how an identical CD8+ T cell product was shaped by melanoma expressing either a low-avidity tumor-associated antigen or high-avidity neoantigen, and kinetically profiled T cell differentiation in these two contexts across host tissues. High-avidity neoantigen expression was sufficient to activate naïve CD8+ T cells - leading to robust tumor regression and long-term protective immunity upon tumor rechallenge. Mechanistically, transferred naïve CD8+ T cells reacting to high-avidity neoantigen exhibited enhanced cytokine production, heightened effector function, and sustained persistence compared to the low-avidity wild-type tumors. Antitumor activity to these high-avidity tumors was preserved even in the absence of functional host T and B lymphocytes, and early lymph node trafficking was found to be essential for ACT efficacy. Expanded effector or stem-memory T cells were compared to the naïve pmel-1 T cell product. Stem-memory but not effector-memory cells exhibited similar antitumor efficacy and lymph node trafficking patterns to the naïve cells in mice with high-avidity neoantigen tumors. These findings highlight how differential tumor antigens shape divergent cellular fate and uncover a critical role of T cell trafficking in lymph nodes in shaping high-avidity neoantigen-specific antitumor responses.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.2,"publicationDate":"2025-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145676656","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 "Function-First" Approach to Identify Regulatory T cell-Targeting Antibodies for Immunotherapy. 鉴定用于免疫治疗的调节性T细胞靶向抗体的“功能优先”方法。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-02 DOI: 10.1158/2326-6066.CIR-24-1083
Kirstie L S Cleary, Monika Semmrich, Linda Martensson, Ulla-Carin Tornberg, David Ermert, Martin C Taylor, Robert J Oldham, Osman Dadas, Josephine F Buckingham, Jinny Kim, H T Claude Chan, Angelica Palm, Ann-Helen Fischer, Mimoza Boden, Jenny Mattsson, Petra Holmkvist, Sean H Lim, Stephen A Beers, Bjorn Frendeus, Ingrid Teige, Mark S Cragg

Despite advances in cancer immunotherapy, treatment response is still highly variable. One contributing factor is the tumor microenvironment and specifically the presence of suppressive immune cells such as regulatory T (Treg) cells. Being able to target these specifically, while leaving effector T-cell populations untouched, is an attractive strategy that may overcome some of these issues, improving responses. To generate antibodies specific for tumor-associated Tregs, lymphocytes were isolated from tumor-bearing mice and panned against the n-CoDeR phage antibody library. Using the target-agnostic F.I.R.S.T. discovery platform, they were evaluated in ex vivo and in vivo models to determine tissue and cell selectivity and specificity and ability to deplete Tregs and elicit tumor control in subcutaneous tumor models. A total of 24 antibodies were identified and explored, representing a range of specificities from pan-T cell to Treg and tumor Treg specific. Relative expression/binding of these mAbs on tumor Tregs was not a predictor of subsequent Treg deletion efficacy or tumor control, whereas tumor Treg selectivity was. One mAb in particular demonstrated tumor-specific depletion of Tregs, leaving those in the spleen and blood untouched. This Fc:FcγR-mediated tumor-specific Treg depletion was important for antitumor effects. Target deconvolution showed that this mAb binds a distinct epitope within ICAM-1, which is hypothesized to mediate its selectivity toward tumor Tregs. These data validate the target-agnostic discovery approach as a viable means to identify new therapeutic antibodies.

尽管癌症免疫疗法取得了进展,但治疗反应仍然是高度可变的。其中一个影响因素是肿瘤微环境,特别是抑制性免疫细胞如调节性T (Treg)细胞的存在。在不影响效应T细胞群的同时,能够特异性地靶向这些细胞是一种有吸引力的策略,可以克服这些问题,改善反应。为了产生针对肿瘤相关Tregs的特异性抗体,从荷瘤小鼠中分离淋巴细胞,并对n-CoDeR噬菌体抗体库进行筛选。利用靶向不可知的F.I.R.S.T.发现平台,在体内和体外模型中对它们进行评估,以确定组织和细胞的选择性和特异性,以及在皮下肿瘤模型中消耗Tregs和引发肿瘤控制的能力。共鉴定和探索了24种抗体,代表了从泛t细胞到Treg和肿瘤Treg特异性的一系列特异性。这些单克隆抗体在肿瘤Treg上的相对表达/结合并不是随后Treg缺失效果或肿瘤控制的预测因子,而肿瘤Treg选择性是。其中一种单克隆抗体特别证明了肿瘤特异性treg的消耗,而脾脏和血液中的treg未受影响。Fc:FcR介导的肿瘤特异性Treg消耗对抗肿瘤作用很重要。靶标反卷积显示,该单抗结合了ICAM-1中一个不同的表位,这被假设介导了其对肿瘤Tregs的选择性。这些数据验证了靶向不可知发现方法作为鉴定新的治疗性抗体的可行手段。
{"title":"A \"Function-First\" Approach to Identify Regulatory T cell-Targeting Antibodies for Immunotherapy.","authors":"Kirstie L S Cleary, Monika Semmrich, Linda Martensson, Ulla-Carin Tornberg, David Ermert, Martin C Taylor, Robert J Oldham, Osman Dadas, Josephine F Buckingham, Jinny Kim, H T Claude Chan, Angelica Palm, Ann-Helen Fischer, Mimoza Boden, Jenny Mattsson, Petra Holmkvist, Sean H Lim, Stephen A Beers, Bjorn Frendeus, Ingrid Teige, Mark S Cragg","doi":"10.1158/2326-6066.CIR-24-1083","DOIUrl":"10.1158/2326-6066.CIR-24-1083","url":null,"abstract":"<p><p>Despite advances in cancer immunotherapy, treatment response is still highly variable. One contributing factor is the tumor microenvironment and specifically the presence of suppressive immune cells such as regulatory T (Treg) cells. Being able to target these specifically, while leaving effector T-cell populations untouched, is an attractive strategy that may overcome some of these issues, improving responses. To generate antibodies specific for tumor-associated Tregs, lymphocytes were isolated from tumor-bearing mice and panned against the n-CoDeR phage antibody library. Using the target-agnostic F.I.R.S.T. discovery platform, they were evaluated in ex vivo and in vivo models to determine tissue and cell selectivity and specificity and ability to deplete Tregs and elicit tumor control in subcutaneous tumor models. A total of 24 antibodies were identified and explored, representing a range of specificities from pan-T cell to Treg and tumor Treg specific. Relative expression/binding of these mAbs on tumor Tregs was not a predictor of subsequent Treg deletion efficacy or tumor control, whereas tumor Treg selectivity was. One mAb in particular demonstrated tumor-specific depletion of Tregs, leaving those in the spleen and blood untouched. This Fc:FcγR-mediated tumor-specific Treg depletion was important for antitumor effects. Target deconvolution showed that this mAb binds a distinct epitope within ICAM-1, which is hypothesized to mediate its selectivity toward tumor Tregs. These data validate the target-agnostic discovery approach as a viable means to identify new therapeutic antibodies.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1969-1986"},"PeriodicalIF":8.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12670074/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145147686","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
Liver Metastases License Neutrophils through IL1 to Potentiate Cancer Progression. 肝转移允许中性粒细胞通过IL-1促进癌症进展。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-02 DOI: 10.1158/2326-6066.CIR-24-1074
Ashley N Pearson, Jessica Waninger, Amanda K Huber, Erin A Holcomb, Jadyn G James, Justine Kyi, Ameer L Elaimy, Zhuwen Wang, Emily L Lasse-Opsahl, Shuvasree SenGupta, David A Elliott, Enid Choi, Qiang Zhang, Meredith A Morgan, Daniel T Chang, Theodore S Lawrence, Adam Courtney, Yatrik M Shah, Jason S Knight, Marina Pasca Di Magliano, Shinjae Yoo, Silvia Crivelli, Carole A Parent, Nithya Ramnath, Alex K Bryant, Weiping Zou, Michael D Green

Liver metastases are associated with poor cancer outcomes in many solid malignancies, but the factors influencing the trajectory of patients with liver metastases are poorly defined. It is known that liver metastases suppress systemic antitumor immunity; however, the underlying mechanisms remain incompletely described. We report that liver metastases promote disease progression in patients and preclinical models. Patients with liver metastases progress rapidly, regardless of primary tumor type. In multiple murine models, we find that liver metastases potentiate neutrophil migration and activity. Neutrophils licensed by liver metastasis augment metastatic colonization in an IL1-dependent manner. Thus, liver metastasis rewires systemic immunity to promote cancer progression. This work has implications for treatment strategies to address the poor clinical outcomes associated with liver metastasis.

在许多实体恶性肿瘤中,肝转移与不良的癌症预后相关,但影响肝转移患者预后的因素尚不明确。众所周知,肝转移抑制全身抗肿瘤免疫;然而,潜在的机制仍然没有完全描述。我们报道肝转移促进患者和临床前模型的疾病进展。无论原发肿瘤类型如何,肝转移患者进展迅速。在多个小鼠模型中,我们发现肝转移增强了中性粒细胞的迁移和活性。嗜中性粒细胞以IL-1依赖的方式增加肝转移的定植。因此,肝转移改变了全身免疫,促进了癌症的进展。这项工作对解决与肝转移相关的不良临床结果的治疗策略具有启示意义。
{"title":"Liver Metastases License Neutrophils through IL1 to Potentiate Cancer Progression.","authors":"Ashley N Pearson, Jessica Waninger, Amanda K Huber, Erin A Holcomb, Jadyn G James, Justine Kyi, Ameer L Elaimy, Zhuwen Wang, Emily L Lasse-Opsahl, Shuvasree SenGupta, David A Elliott, Enid Choi, Qiang Zhang, Meredith A Morgan, Daniel T Chang, Theodore S Lawrence, Adam Courtney, Yatrik M Shah, Jason S Knight, Marina Pasca Di Magliano, Shinjae Yoo, Silvia Crivelli, Carole A Parent, Nithya Ramnath, Alex K Bryant, Weiping Zou, Michael D Green","doi":"10.1158/2326-6066.CIR-24-1074","DOIUrl":"10.1158/2326-6066.CIR-24-1074","url":null,"abstract":"<p><p>Liver metastases are associated with poor cancer outcomes in many solid malignancies, but the factors influencing the trajectory of patients with liver metastases are poorly defined. It is known that liver metastases suppress systemic antitumor immunity; however, the underlying mechanisms remain incompletely described. We report that liver metastases promote disease progression in patients and preclinical models. Patients with liver metastases progress rapidly, regardless of primary tumor type. In multiple murine models, we find that liver metastases potentiate neutrophil migration and activity. Neutrophils licensed by liver metastasis augment metastatic colonization in an IL1-dependent manner. Thus, liver metastasis rewires systemic immunity to promote cancer progression. This work has implications for treatment strategies to address the poor clinical outcomes associated with liver metastasis.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"2023-2036"},"PeriodicalIF":8.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144999749","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: Article Recommendations from Our Deputy and Senior Editors. 从文献的亮点抽样:文章推荐从我们的副和高级编辑。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-02 DOI: 10.1158/2326-6066.CIR-13-12-WWR
{"title":"A Sampling of Highlights from the Literature: Article Recommendations from Our Deputy and Senior Editors.","authors":"","doi":"10.1158/2326-6066.CIR-13-12-WWR","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-13-12-WWR","url":null,"abstract":"","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":"13 12","pages":"1893"},"PeriodicalIF":8.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145653584","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
CD206+CD14- Skin-Resident Macrophages and DC-T Cell Clusters Are Spatial Features Characterizing Nonrelapsing Cutaneous Squamous Cell Carcinoma. CD206+CD14-皮肤巨噬细胞和DC-T细胞簇是非复发性皮肤鳞状细胞癌的空间特征。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-02 DOI: 10.1158/2326-6066.CIR-25-0182
Roxane Elaldi, Aïda Meghraoui, Axel Elaldi, Luciana Petti, Alizé Gouleau, Patrice Hemon, Wassila Khatir, Jonas Meziane, Xavier Descombes, Henri Montaudié, Julien Boyer, Alexandre Bozec, Gilles Poissonnet, Jacques-Olivier Pers, Anne Sudaka, Véronique M Braud, Fabienne Anjuère

Current histopathologic classifications do not reliably distinguish patients with primary cutaneous squamous cell carcinomas (cSCC) at risk of relapse from those with nonrelapsing tumors. This underscores the need for molecular signatures capable of stratifying patients during primary tumor resection. In this study, we used high-dimensional imaging mass cytometry and a 39-antibody panel to define the immune landscape of 20 primary cSCC with distinct clinical outcomes, four relapsing cSCC, and their perilesional skins. Computational analysis of spatially resolved single-cell data from 47 imaging mass cytometry images identified 12 immune-cell subsets that discriminated primary cSCC from perilesional skin. Regulatory T cells, cytotoxic CD8+ T lymphocytes, and tumor-associated macrophages and neutrophils characterized tumors, whereas Langerhans cells and skin-resident macrophages defined perilesional skin. Skin-resident macrophages were characterized by the expression of CD206, CD11c, and HLA-DR and the absence of CD14. These cells infiltrated tumors from nonrelapsing patients more efficiently. We found a higher density of proliferating, mature, and cytotoxic cells within this macrophage subset, consistent with the absence of relapse. Spectral flow cytometry analysis on fresh tumor biopsies revealed that the skin-resident macrophages had phagocytic properties, suggesting a role in tumor antigen processing. Additionally, neighborhood profiling revealed that DC-LAMP+ dendritic cells were in close proximity with helper and cytotoxic T lymphocytes in primary cSCC from patients without relapse, indicative of active adaptive immunity. Our findings identify phagocytic skin-resident macrophages and dendritic cell-T cell clusters as features differentiating nonrelapsing cSCC from primary cSCC at risk of relapse. These data have the potential to guide the identification of prognostic biomarkers for cSCC.

目前的组织病理学分类不能可靠地区分有复发风险的原发性皮肤鳞状细胞癌(cSCC)患者和非复发肿瘤患者。这强调了在原发肿瘤切除期间能够对患者进行分层的分子标记的必要性。在这里,我们使用高维成像细胞计数(IMC)和39抗体小组来定义20例具有不同临床结果的原发性cSCC, 4例复发性cSCC及其病灶周围皮肤的免疫景观。对来自47张IMC图像的空间分辨单细胞数据进行计算分析,确定了12个免疫细胞亚群,可以区分原发性cSCC和病灶周围皮肤。调节性T细胞、细胞毒性CD8+ T淋巴细胞、肿瘤相关巨噬细胞和中性粒细胞是肿瘤的特征,而朗格汉斯细胞和皮肤常驻巨噬细胞是病灶周围皮肤的特征。皮肤巨噬细胞的特征是表达CD206、CD11c和HLA-DR,而不表达CD14。这些细胞更有效地浸润非复发患者的肿瘤。我们发现在这个巨噬细胞亚群中有更高密度的增殖、成熟和细胞毒性细胞,这与没有复发相一致。新鲜肿瘤活检的流式细胞术分析显示,皮肤巨噬细胞具有吞噬特性,提示其在肿瘤抗原加工中起作用。此外,邻域分析显示DC- lamp +树突状细胞(DC)与无复发的原发性cSCC患者的辅助T淋巴细胞和细胞毒性T淋巴细胞非常接近,表明存在主动适应性免疫。我们的研究结果确定了吞噬性皮肤巨噬细胞和DC-T细胞簇是区分复发风险的原发性cSCC和非复发性cSCC的特征。这些数据有可能指导鉴别cSCC的预后生物标志物。
{"title":"CD206+CD14- Skin-Resident Macrophages and DC-T Cell Clusters Are Spatial Features Characterizing Nonrelapsing Cutaneous Squamous Cell Carcinoma.","authors":"Roxane Elaldi, Aïda Meghraoui, Axel Elaldi, Luciana Petti, Alizé Gouleau, Patrice Hemon, Wassila Khatir, Jonas Meziane, Xavier Descombes, Henri Montaudié, Julien Boyer, Alexandre Bozec, Gilles Poissonnet, Jacques-Olivier Pers, Anne Sudaka, Véronique M Braud, Fabienne Anjuère","doi":"10.1158/2326-6066.CIR-25-0182","DOIUrl":"10.1158/2326-6066.CIR-25-0182","url":null,"abstract":"<p><p>Current histopathologic classifications do not reliably distinguish patients with primary cutaneous squamous cell carcinomas (cSCC) at risk of relapse from those with nonrelapsing tumors. This underscores the need for molecular signatures capable of stratifying patients during primary tumor resection. In this study, we used high-dimensional imaging mass cytometry and a 39-antibody panel to define the immune landscape of 20 primary cSCC with distinct clinical outcomes, four relapsing cSCC, and their perilesional skins. Computational analysis of spatially resolved single-cell data from 47 imaging mass cytometry images identified 12 immune-cell subsets that discriminated primary cSCC from perilesional skin. Regulatory T cells, cytotoxic CD8+ T lymphocytes, and tumor-associated macrophages and neutrophils characterized tumors, whereas Langerhans cells and skin-resident macrophages defined perilesional skin. Skin-resident macrophages were characterized by the expression of CD206, CD11c, and HLA-DR and the absence of CD14. These cells infiltrated tumors from nonrelapsing patients more efficiently. We found a higher density of proliferating, mature, and cytotoxic cells within this macrophage subset, consistent with the absence of relapse. Spectral flow cytometry analysis on fresh tumor biopsies revealed that the skin-resident macrophages had phagocytic properties, suggesting a role in tumor antigen processing. Additionally, neighborhood profiling revealed that DC-LAMP+ dendritic cells were in close proximity with helper and cytotoxic T lymphocytes in primary cSCC from patients without relapse, indicative of active adaptive immunity. Our findings identify phagocytic skin-resident macrophages and dendritic cell-T cell clusters as features differentiating nonrelapsing cSCC from primary cSCC at risk of relapse. These data have the potential to guide the identification of prognostic biomarkers for cSCC.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1925-1937"},"PeriodicalIF":8.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145147736","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
BFAR Promotes Neutrophil Infiltration and Immunosuppressive Reprogramming through the PRP19-YBX1 Axis to Induce Immune Evasion in Gastric Cancer. BFAR通过PRP19-YBX1轴促进中性粒细胞浸润和免疫抑制重编程,诱导胃癌免疫逃避。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-02 DOI: 10.1158/2326-6066.CIR-24-1011
Xin Ma, Yumei Liu, Yingying Chen, Juan Wang, Feiyue Zhang, Wei Liang, Pengbo Zhang, Yunlan Zhou, Bei Miao, Sujuan Fei, Masami Yamamoto, Tetsuya Tsukamoto, Sachiyo Nomura, Li Li, Jiajia Wang

The immunosuppressive tumor microenvironment remains a major barrier to effective immunotherapy in gastric cancer. In this study, we identified the E3 ubiquitin ligase BFAR as a critical regulator of neutrophil-mediated immune evasion through the S100A8/A9-BFAR-PRP19-YBX1 signaling axis. Multiomics analyses revealed that BFAR is overexpressed in gastric cancer and correlates with poor prognosis. Functional studies demonstrated that BFAR knockdown suppressed tumor growth by reducing neutrophil infiltration and immunosuppressive reprogramming to restore CD8+ T-cell function. Mechanistically, BFAR mediated K48-linked ubiquitination and degradation of PRP19, leading to stabilization of the oncoprotein YBX1, which transcriptionally upregulated neutrophil-recruiting chemokines CXCL1/CXCL3. Infiltrating neutrophils secreted S100A8/A9, which activated NF-κB to induce BFAR expression in tumor cells and created a feed-forward loop that sustains an immunosuppressive tumor microenvironment. Furthermore, BFAR promoted neutrophil PD-L1 expression via GM-CSF, reinforcing T-cell exhaustion. Clinically, BFAR expression correlated with neutrophil infiltration and poor response to anti-PD-1 therapy, whereas its inhibition synergizes with immune checkpoint blockade in preclinical models. Our work unveils BFAR as a central orchestrator of neutrophil-driven immunosuppression and proposes targeting this axis to enhance immunotherapy efficacy in gastric cancer.

免疫抑制肿瘤微环境(TME)仍然是胃癌(GC)有效免疫治疗的主要障碍。在这里,我们发现E3泛素连接酶BFAR是通过S100A8/A9-BFAR-PRP19-YBX1信号轴介导中性粒细胞介导的免疫逃避的关键调节因子。多组学分析显示BFAR在胃癌中过表达,与不良预后相关。功能研究表明,BFAR敲低通过减少中性粒细胞浸润和免疫抑制重编程来恢复CD8+ T细胞功能,从而抑制肿瘤生长。在机制上,BFAR介导k48相关的泛素化和PRP19的降解,导致癌蛋白YBX1的稳定,从而上调中性粒细胞募集趋化因子CXCL1/CXCL3的转录。浸润的中性粒细胞分泌S100A8/A9,激活NF-κB诱导肿瘤细胞中BFAR的表达,形成前馈循环,维持免疫抑制TME。此外,BFAR通过GM-CSF促进中性粒细胞PD-L1表达,增强T细胞衰竭。在临床上,BFAR的表达与中性粒细胞浸润和对抗pd -1治疗的不良反应相关,而在临床前模型中,其抑制作用与免疫检查点阻断具有协同作用。我们的工作揭示了BFAR是中性粒细胞驱动的免疫抑制的中心协调者,并提出了靶向该轴以提高GC的免疫治疗效果。
{"title":"BFAR Promotes Neutrophil Infiltration and Immunosuppressive Reprogramming through the PRP19-YBX1 Axis to Induce Immune Evasion in Gastric Cancer.","authors":"Xin Ma, Yumei Liu, Yingying Chen, Juan Wang, Feiyue Zhang, Wei Liang, Pengbo Zhang, Yunlan Zhou, Bei Miao, Sujuan Fei, Masami Yamamoto, Tetsuya Tsukamoto, Sachiyo Nomura, Li Li, Jiajia Wang","doi":"10.1158/2326-6066.CIR-24-1011","DOIUrl":"10.1158/2326-6066.CIR-24-1011","url":null,"abstract":"<p><p>The immunosuppressive tumor microenvironment remains a major barrier to effective immunotherapy in gastric cancer. In this study, we identified the E3 ubiquitin ligase BFAR as a critical regulator of neutrophil-mediated immune evasion through the S100A8/A9-BFAR-PRP19-YBX1 signaling axis. Multiomics analyses revealed that BFAR is overexpressed in gastric cancer and correlates with poor prognosis. Functional studies demonstrated that BFAR knockdown suppressed tumor growth by reducing neutrophil infiltration and immunosuppressive reprogramming to restore CD8+ T-cell function. Mechanistically, BFAR mediated K48-linked ubiquitination and degradation of PRP19, leading to stabilization of the oncoprotein YBX1, which transcriptionally upregulated neutrophil-recruiting chemokines CXCL1/CXCL3. Infiltrating neutrophils secreted S100A8/A9, which activated NF-κB to induce BFAR expression in tumor cells and created a feed-forward loop that sustains an immunosuppressive tumor microenvironment. Furthermore, BFAR promoted neutrophil PD-L1 expression via GM-CSF, reinforcing T-cell exhaustion. Clinically, BFAR expression correlated with neutrophil infiltration and poor response to anti-PD-1 therapy, whereas its inhibition synergizes with immune checkpoint blockade in preclinical models. Our work unveils BFAR as a central orchestrator of neutrophil-driven immunosuppression and proposes targeting this axis to enhance immunotherapy efficacy in gastric cancer.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"2037-2053"},"PeriodicalIF":8.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12670071/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145085127","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
Antibody-Mediated Inhibition of HLA/LILR Interactions Breaks Innate Immune Tolerance and Induces Antitumor Immunity. 抗体介导的HLA/LILR相互作用抑制打破先天免疫耐受并诱导抗肿瘤免疫。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-02 DOI: 10.1158/2326-6066.CIR-25-0343
Abir K Panda, Kannan Natarajan, Surajit Sinha, Jiansheng Jiang, Sruthi Chempati, Lisa F Boyd, Priyanka P Desai, Maja Buszko, Yong-Hee Kim, Soha Kazmi, Bryan Fisk, Martha E Teke, Carolina M Larrain, Kirsten Remmert, Andrew M Blakely, Iyadh Douagi, Jonathan M Hernandez, David H Margulies, Ethan M Shevach

Immune checkpoint blockade for the treatment of malignancies has been focused on reversing inhibitory pathways in T lymphocytes. NK cells are a potent innate defense against tumors and virally infected cells, but their therapeutic manipulation for anticancer immunity has been inadequately explored. Considerable attention has been focused on approaches to blocking inhibitory receptors on NK and myeloid cells. Most effort has been directed to the killer immunoglobulin-like receptors and CD94/NKG2A on NK cells. Another set of receptors with similar function in both NK cells and myeloid cells is the leukocyte immunoglobulin-like receptors (LILR) that interact with a wide variety of HLA molecules. Using pan-anti-HLA mAbs that recognize a conserved epitopic region on HLA also seen by LILRs, we investigated their functional effects in several models of tumor immunity. The pan-anti-HLA mAbs blocked the binding of most LILRs and did not block killer cell immunoglobulin-like receptors or CD94/NKG2A/C or T-cell receptor recognition. They also activated dysfunctional NK cells explanted from a variety of human cancers and resulted in enhancement of tumor immunity in humanized mice. The mAbs also exert direct antitumor effects. These results suggest that activation of innate immunity via disruption of HLA/LILR interactions is a potent approach for control of both primary tumors and potentially tumor metastases.

免疫检查点阻断治疗恶性肿瘤的重点是逆转T淋巴细胞的抑制途径。自然杀伤(NK)细胞是对抗肿瘤和病毒感染细胞的一种有效的先天防御,但其在抗癌免疫中的治疗作用尚未得到充分的探讨。大量的注意力集中在阻断NK细胞和髓细胞上的抑制性受体的方法上。大多数研究都是针对NK细胞上的杀伤性免疫球蛋白样受体(KIR)和CD94/NKG2A。另一组在NK细胞和髓细胞中具有类似功能的受体是白细胞免疫球蛋白样受体(LILR),它与多种HLA分子相互作用。利用泛抗HLA单克隆抗体,我们研究了它们在几种肿瘤免疫模型中的功能作用,这些单克隆抗体识别HLA上的保守表位区域,也被LILR所看到。泛抗hla单抗阻断大多数lilr的结合,不阻断杀伤细胞免疫球蛋白样受体(KIR)或CD94/NKG2A/C或TCR识别。它们还激活了从多种人类癌症中移植的功能失调的NK细胞,并导致人源化小鼠的肿瘤免疫增强。单克隆抗体还具有直接的抗肿瘤作用。这些结果表明,通过破坏HLA/LILR相互作用激活先天免疫是控制原发性肿瘤和潜在肿瘤转移的有效途径。
{"title":"Antibody-Mediated Inhibition of HLA/LILR Interactions Breaks Innate Immune Tolerance and Induces Antitumor Immunity.","authors":"Abir K Panda, Kannan Natarajan, Surajit Sinha, Jiansheng Jiang, Sruthi Chempati, Lisa F Boyd, Priyanka P Desai, Maja Buszko, Yong-Hee Kim, Soha Kazmi, Bryan Fisk, Martha E Teke, Carolina M Larrain, Kirsten Remmert, Andrew M Blakely, Iyadh Douagi, Jonathan M Hernandez, David H Margulies, Ethan M Shevach","doi":"10.1158/2326-6066.CIR-25-0343","DOIUrl":"10.1158/2326-6066.CIR-25-0343","url":null,"abstract":"<p><p>Immune checkpoint blockade for the treatment of malignancies has been focused on reversing inhibitory pathways in T lymphocytes. NK cells are a potent innate defense against tumors and virally infected cells, but their therapeutic manipulation for anticancer immunity has been inadequately explored. Considerable attention has been focused on approaches to blocking inhibitory receptors on NK and myeloid cells. Most effort has been directed to the killer immunoglobulin-like receptors and CD94/NKG2A on NK cells. Another set of receptors with similar function in both NK cells and myeloid cells is the leukocyte immunoglobulin-like receptors (LILR) that interact with a wide variety of HLA molecules. Using pan-anti-HLA mAbs that recognize a conserved epitopic region on HLA also seen by LILRs, we investigated their functional effects in several models of tumor immunity. The pan-anti-HLA mAbs blocked the binding of most LILRs and did not block killer cell immunoglobulin-like receptors or CD94/NKG2A/C or T-cell receptor recognition. They also activated dysfunctional NK cells explanted from a variety of human cancers and resulted in enhancement of tumor immunity in humanized mice. The mAbs also exert direct antitumor effects. These results suggest that activation of innate immunity via disruption of HLA/LILR interactions is a potent approach for control of both primary tumors and potentially tumor metastases.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1938-1955"},"PeriodicalIF":8.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12560147/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145198503","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
Spatiotemporal Immune Landscape and Long-term Immune Memory in POLE-Mutant Endometrial Cancer at the Single-Cell Level. 在单细胞水平上pole突变子宫内膜癌的时空免疫景观和长期免疫记忆。
IF 8.2 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-12-02 DOI: 10.1158/2326-6066.CIR-25-0083
Koen Brummel, Marta Requesens, Nienke van Rooij, Hagma H Workel, Florine A Eggink, Annechien Plat, René Wardenaar, Diana C J Spierings, Floris Foijer, David N Church, Joost Bart, Hans W Nijman, Marco de Bruyn

Polymerase epsilon-mutant (POLE-mut) endometrial cancers are characterized by a near 100% disease-specific survival rate, even when treated with surgery alone. This survival, combined with the ultramutated genome and high level of neoantigens in these tumors, indicates a substantial degree of immune control in preventing disease spread and recurrence. Although these features are intriguing, the analysis of immune infiltration in POLE-mut endometrial cancers has predominantly been confined to IHC studies. In this study, we used single-cell RNA and T-cell receptor sequencing to characterize the immune landscape of POLE-mut endometrial cancers. Moreover, we analyzed patient blood samples taken 2 to 8 years after curative treatment to assess the formation of long-term immune memory in circulation. We identified specialized tumor-infiltrating myeloid subsets at different stages of maturation, an array of lymphocytes ranging from immature to cytotoxic, and adaptive NK cells, as well as tumor-reactive exhausted and effector T cells, all contributing to a highly inflammatory antitumor response. Our analysis of blood samples taken years after curative treatment uncovered the presence of tumor-reactive T-cell clones that matched the primary tumor. This indicates the formation of systemic long-term memory immune responses in POLE-mut endometrial cancer survivors. Our study highlights the distinctive immunogenicity of POLE-mut endometrial cancer and identifies key features associated with persistent antitumor immunity that may contribute to prolonged, relapse-free survival.

聚合酶epsilon突变(POLE-mut)子宫内膜癌(EC)的特点是接近100%的疾病特异性生存率,即使仅通过手术治疗。这种惊人的存活率,加上这些肿瘤中超突变的基因组和高水平的新抗原,表明在预防疾病扩散和复发方面有相当程度的免疫控制。尽管这些特征很有趣,但POLE-mut EC的免疫浸润主要局限于免疫组织化学研究。在这里,我们使用最先进的单细胞RNA和TCR测序来表征pole突变ec的免疫景观。此外,我们独特地分析了治愈治疗后2至8年的患者血液样本,以评估循环中长期免疫记忆的形成。我们确定了不同成熟阶段的特殊肿瘤浸润骨髓亚群,一系列淋巴细胞,从未成熟到细胞毒性和适应性自然杀伤(NK),以及肿瘤反应性耗尽和效应T细胞,有助于高度炎症抗肿瘤反应。值得注意的是,我们对治疗后数年采集的血液样本进行分析,发现了与原发肿瘤相匹配的肿瘤反应性T细胞克隆的存在。这表明在POLE-mut EC幸存者中形成了系统性长期记忆免疫反应。我们的研究强调了POLE-mut EC独特的免疫原性,并确定了与持续抗肿瘤免疫相关的关键特征,这可能有助于延长无复发生存期。
{"title":"Spatiotemporal Immune Landscape and Long-term Immune Memory in POLE-Mutant Endometrial Cancer at the Single-Cell Level.","authors":"Koen Brummel, Marta Requesens, Nienke van Rooij, Hagma H Workel, Florine A Eggink, Annechien Plat, René Wardenaar, Diana C J Spierings, Floris Foijer, David N Church, Joost Bart, Hans W Nijman, Marco de Bruyn","doi":"10.1158/2326-6066.CIR-25-0083","DOIUrl":"10.1158/2326-6066.CIR-25-0083","url":null,"abstract":"<p><p>Polymerase epsilon-mutant (POLE-mut) endometrial cancers are characterized by a near 100% disease-specific survival rate, even when treated with surgery alone. This survival, combined with the ultramutated genome and high level of neoantigens in these tumors, indicates a substantial degree of immune control in preventing disease spread and recurrence. Although these features are intriguing, the analysis of immune infiltration in POLE-mut endometrial cancers has predominantly been confined to IHC studies. In this study, we used single-cell RNA and T-cell receptor sequencing to characterize the immune landscape of POLE-mut endometrial cancers. Moreover, we analyzed patient blood samples taken 2 to 8 years after curative treatment to assess the formation of long-term immune memory in circulation. We identified specialized tumor-infiltrating myeloid subsets at different stages of maturation, an array of lymphocytes ranging from immature to cytotoxic, and adaptive NK cells, as well as tumor-reactive exhausted and effector T cells, all contributing to a highly inflammatory antitumor response. Our analysis of blood samples taken years after curative treatment uncovered the presence of tumor-reactive T-cell clones that matched the primary tumor. This indicates the formation of systemic long-term memory immune responses in POLE-mut endometrial cancer survivors. Our study highlights the distinctive immunogenicity of POLE-mut endometrial cancer and identifies key features associated with persistent antitumor immunity that may contribute to prolonged, relapse-free survival.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1911-1924"},"PeriodicalIF":8.2,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144944001","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:604180095
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