Cancer immunology research最新文献

筛选
英文 中文
The HSP90 Inhibitor Pimitespib Targets Regulatory T Cells in the Tumor Microenvironment. HSP90抑制剂Pimitespib靶向肿瘤微环境中的调节性T细胞
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2025-02-03 DOI: 10.1158/2326-6066.CIR-24-0713
Ayaka Tsuge, Sho Watanabe, Akihito Kawazoe, Yosuke Togashi, Kota Itahashi, Mari Masuda, Atsuo Sai, Shogo Takei, Hiromi Muraoka, Shuichi Ohkubo, Daisuke Sugiyama, Yue Yan, Shota Fukuoka, Toshihiko Doi, Kohei Shitara, Shohei Koyama, Hiroyoshi Nishikawa
{"title":"The HSP90 Inhibitor Pimitespib Targets Regulatory T Cells in the Tumor Microenvironment.","authors":"Ayaka Tsuge, Sho Watanabe, Akihito Kawazoe, Yosuke Togashi, Kota Itahashi, Mari Masuda, Atsuo Sai, Shogo Takei, Hiromi Muraoka, Shuichi Ohkubo, Daisuke Sugiyama, Yue Yan, Shota Fukuoka, Toshihiko Doi, Kohei Shitara, Shohei Koyama, Hiroyoshi Nishikawa","doi":"10.1158/2326-6066.CIR-24-0713","DOIUrl":"10.1158/2326-6066.CIR-24-0713","url":null,"abstract":"<p><p>Regulatory T (Treg) cells play key roles in cancer immunity by suppressing a range of antitumor immune responses and contributing to resistance to PD-1 blockade therapy. Given their critical roles in self-tolerance, local control of immunosuppression by Treg cells, such as in the tumor microenvironment, has been intensively studied. Inhibition of HSP90, a chaperone with vital roles in regulating proteostasis in cancer cells, impedes cancer progression by interrupting oncogenic signaling pathways and potentially modulating antitumor immunity, but we have very little mechanistic insight into these immune modulatory effects. In this study, we show that the number of Treg cells is selectively reduced by the HSP90 inhibitor pimitespib in animal models and patients with gastric cancer in a clinical trial (EPOC1704). Pimitespib reduced the highly immunosuppressive human FOXP3high effector Treg cells by inhibiting their proliferation and decreasing their expression of effector molecules, which improved the priming and activation of antigen-specific CD8+ T cells. Mechanistic studies revealed that pimitespib selectively degraded STAT5, a key transducer of the IL2 signaling pathway, which is essential for Treg cell development and maintenance, and consequently compromised FOXP3 expression, leading to selective impairment of immunosuppression in the tumor microenvironment by Treg cells. Thus, pimitespib treatment combined with PD-1 blockade exhibited a far stronger antitumor effect than either treatment alone in animal models. Through these data, we propose that HSP90 inhibition is a promising therapeutic option for Treg cell-targeted cancer immunotherapy.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"273-285"},"PeriodicalIF":8.1,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142738522","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
Identification and Phenotypic Characterization of Neoantigen-Specific Cytotoxic CD4+ T Cells in Endometrial Cancer. 子宫内膜癌中新抗原特异性细胞毒性CD4+ T细胞的鉴定和表型特征。
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2025-02-03 DOI: 10.1158/2326-6066.CIR-24-0514
Minami Fusagawa, Serina Tokita, Kenji Murata, Tasuku Mariya, Mina Umemoto, Shintaro Sugita, Kazuhiko Matsuo, Yoshihiko Hirohashi, Tsuyoshi Saito, Takayuki Kanaseki, Toshihiko Torigoe
{"title":"Identification and Phenotypic Characterization of Neoantigen-Specific Cytotoxic CD4+ T Cells in Endometrial Cancer.","authors":"Minami Fusagawa, Serina Tokita, Kenji Murata, Tasuku Mariya, Mina Umemoto, Shintaro Sugita, Kazuhiko Matsuo, Yoshihiko Hirohashi, Tsuyoshi Saito, Takayuki Kanaseki, Toshihiko Torigoe","doi":"10.1158/2326-6066.CIR-24-0514","DOIUrl":"10.1158/2326-6066.CIR-24-0514","url":null,"abstract":"<p><p>Tumor-reactive CD4+ T cells often accumulate in the tumor microenvironment (TME) in human cancer, but their functions and roles in antitumor responses remain elusive. Here, we investigated the immunopeptidome of HLA class II-positive (HLA-II+) endometrial cancer with an inflamed TME using a proteogenomic approach. We identified HLA-II neoantigens, one of which induced polyclonal CD4+ tumor-infiltrating lymphocyte responses. We then experimentally demonstrated that neoantigen-specific CD4+ tumor-infiltrating lymphocytes lyse target cells in an HLA-II-dependent manner. Single-cell transcriptomic analysis of the TME coupled with T-cell receptor sequencing revealed the presence of CD4+ T-cell clusters characterized by CXCL13 expression. The CXCL13+ clusters contained two subclusters with distinct cytotoxic gene expression patterns. The identified neoantigen-specific CD4+ T cells were found exclusively in one of the CXCL13+ subclusters characterized by granzyme B and CCL5 expression. These results demonstrate the involvement of tumor-reactive CD4+ T cells with cytotoxic function in immune surveillance of endometrial cancer and reveal their transcriptomic signature.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"171-184"},"PeriodicalIF":8.1,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142799403","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
TBK1 Targeting Is Identified as a Therapeutic Strategy to Enhance CAR T-Cell Efficacy Using Patient-Derived Organotypic Tumor Spheroids. TBK1靶向被认为是一种利用患者来源的器官型肿瘤球体增强CAR - t细胞疗效的治疗策略。
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2025-02-03 DOI: 10.1158/2326-6066.CIR-23-1011
Yi Sun, Luke Maggs, Apekshya Panda, Samuel J Wright, Angelina M Cicerchia, Anne Jenney, Matthew D Perricone, Caitlin E Mills, Giulia Cattaneo, Marco Ventin, Feng Chen, Martin Q Rasmussen, Alex Miranda, Or-Yam Revach, Jacy Fang, Amina Fu, Peter J Bowling, Tatyana Sharova, Aleigha Lawless, Peter K Sorger, Nabeel Bardeesy, Xinhui Wang, Keith T Flaherty, Genevieve M Boland, Arnav Mehta, Moshe Sade-Feldman, Cristina R Ferrone, Russell W Jenkins
{"title":"TBK1 Targeting Is Identified as a Therapeutic Strategy to Enhance CAR T-Cell Efficacy Using Patient-Derived Organotypic Tumor Spheroids.","authors":"Yi Sun, Luke Maggs, Apekshya Panda, Samuel J Wright, Angelina M Cicerchia, Anne Jenney, Matthew D Perricone, Caitlin E Mills, Giulia Cattaneo, Marco Ventin, Feng Chen, Martin Q Rasmussen, Alex Miranda, Or-Yam Revach, Jacy Fang, Amina Fu, Peter J Bowling, Tatyana Sharova, Aleigha Lawless, Peter K Sorger, Nabeel Bardeesy, Xinhui Wang, Keith T Flaherty, Genevieve M Boland, Arnav Mehta, Moshe Sade-Feldman, Cristina R Ferrone, Russell W Jenkins","doi":"10.1158/2326-6066.CIR-23-1011","DOIUrl":"10.1158/2326-6066.CIR-23-1011","url":null,"abstract":"<p><p>Novel therapeutic strategies are needed to improve the efficacy of chimeric antigen receptor (CAR) T cells as a treatment of solid tumors. Multiple tumor microenvironmental factors are thought to contribute to resistance to CAR T-cell therapy in solid tumors, and appropriate model systems to identify and examine these factors using clinically relevant biospecimens are limited. In this study, we examined the activity of B7-H3-directed CAR T cells (B7-H3.CAR-T) using 3D microfluidic cultures of patient-derived organotypic tumor spheroids (PDOTS) and then confirmed the activity of B7-H3.CAR T cells in PDOTS. Although B7-H3 expression in PDOTS was associated with B7-H3.CAR-T sensitivity, mechanistic studies revealed dynamic upregulation of co-inhibitory receptors on CAR T-cells following target cell encounter that led to CAR T-cell dysfunction and limited efficacy against B7-H3-expressing tumors. PD-1 blockade restored CAR T-cell activity in monotypic and organotypic tumor spheroids with improved tumor control and upregulation of effector cytokines. Given the emerging role of TANK-binding kinase 1 (TBK1) as an immune evasion gene, we examined the effect of TBK1 inhibition on CAR T-cell efficacy. Similar to PD-1 blockade, TBK1 inhibition restored CAR T-cell activity in monotypic and organotypic tumor spheroids, prevented CAR T-cell dysfunction, and enhanced CAR T-cell proliferation. Inhibition or deletion of TBK1 also enhanced the sensitivity of cancer cells to immune-mediated killing. Taken together, our results demonstrate the feasibility and utility of ex vivo profiling of CAR T cells using PDOTS and suggest that targeting TBK1 could be used to enhance CAR T-cell efficacy by overcoming tumor-intrinsic and -extrinsic resistance mechanisms.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"210-228"},"PeriodicalIF":8.1,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142945092","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
Hyperthermic Intrathoracic Chemotherapy Modulates the Immune Microenvironment of Pleural Mesothelioma and Improves the Impact of Dual Immune Checkpoint Inhibition. 胸腔内热化疗可调节胸膜间皮瘤的免疫微环境,并改善双重免疫检查点抑制的效果。
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2025-02-03 DOI: 10.1158/2326-6066.CIR-24-0245
Yameng Hao, Aspasia Gkasti, Amy J Managh, Julien Dagher, Alexandros Sifis, Luca Tiron, Louis-Emmanuel Chriqui, Damien N Marie, Olga De Souza Silva, Michel Christodoulou, Solange Peters, Johanna A Joyce, Thorsten Krueger, Michel Gonzalez, Etienne Abdelnour-Berchtold, Christine Sempoux, Daniel Clerc, Hugo Teixeira-Farinha, Martin Hübner, Etienne Meylan, Paul J Dyson, Sabrina Cavin, Jean Y Perentes
{"title":"Hyperthermic Intrathoracic Chemotherapy Modulates the Immune Microenvironment of Pleural Mesothelioma and Improves the Impact of Dual Immune Checkpoint Inhibition.","authors":"Yameng Hao, Aspasia Gkasti, Amy J Managh, Julien Dagher, Alexandros Sifis, Luca Tiron, Louis-Emmanuel Chriqui, Damien N Marie, Olga De Souza Silva, Michel Christodoulou, Solange Peters, Johanna A Joyce, Thorsten Krueger, Michel Gonzalez, Etienne Abdelnour-Berchtold, Christine Sempoux, Daniel Clerc, Hugo Teixeira-Farinha, Martin Hübner, Etienne Meylan, Paul J Dyson, Sabrina Cavin, Jean Y Perentes","doi":"10.1158/2326-6066.CIR-24-0245","DOIUrl":"10.1158/2326-6066.CIR-24-0245","url":null,"abstract":"<p><p>Pleural mesothelioma is a fatal disease with limited treatment options. Recently, pleural mesothelioma management has improved with the development of immune checkpoint inhibitors (ICI). In first-line therapy, dual PD-1 and CTLA-4 blockade enhances tumor control and patient survival compared with chemotherapy. Unfortunately, only a fraction of patients is responsive to immunotherapy, and approaches to reshape the tumor immune microenvironment and make ICIs more effective are urgently required. In this study, we evaluated the effect of hyperthermic intrathoracic chemotherapy (HITOC), a treatment that combines fever-range hyperthermia with local intrapleural cisplatin chemotherapy, on the tumor immune microenvironment and response to ICIs. To do this, we developed a murine pleural mesothelioma model of HITOC. We found that HITOC significantly improved tumor control and animal survival through a mechanism involving the development of a cytotoxic immune response. Additionally, HITOC enhanced immune checkpoint expression by T lymphocytes and synergized with dual PD-1 and CTLA-4 inhibition, leading to further improvement in animal survival. Finally, the analysis of peritoneal mesothelioma patient samples treated by pressurized intraperitoneal aerosol chemotherapy revealed a similar immunomodulation. In conclusion, HITOC remodels the tumor immune microenvironment of pleural mesothelioma by promoting T-cell infiltration into the tumor and could be considered in combination with ICIs in the context of a clinical trial.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"185-199"},"PeriodicalIF":8.1,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142709128","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting of Tumoral NAC1 Mitigates Myeloid-Derived Suppressor Cell-Mediated Immunosuppression and Potentiates Anti-PD-1 Therapy in Ovarian Cancer. 靶向肿瘤 NAC1 可减轻髓源性抑制细胞介导的免疫抑制,并增强卵巢癌的抗 PD-1 治疗效果。
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2025-02-03 DOI: 10.1158/2326-6066.CIR-24-0084
Shunli Dong, Cong Ye, Bin Li, Fanglin Lv, Lu Zhang, Shumin Yang, Fang Wang, Mingxian Zhu, Mingxuan Zhou, Fanfan Guo, Zhenyun Li, Lei Peng, Cheng Ji, Xialiang Lu, Yan Cheng, Xingcong Ren, Youguo Chen, Jinhua Zhou, Jinming Yang, Yi Zhang
{"title":"Targeting of Tumoral NAC1 Mitigates Myeloid-Derived Suppressor Cell-Mediated Immunosuppression and Potentiates Anti-PD-1 Therapy in Ovarian Cancer.","authors":"Shunli Dong, Cong Ye, Bin Li, Fanglin Lv, Lu Zhang, Shumin Yang, Fang Wang, Mingxian Zhu, Mingxuan Zhou, Fanfan Guo, Zhenyun Li, Lei Peng, Cheng Ji, Xialiang Lu, Yan Cheng, Xingcong Ren, Youguo Chen, Jinhua Zhou, Jinming Yang, Yi Zhang","doi":"10.1158/2326-6066.CIR-24-0084","DOIUrl":"10.1158/2326-6066.CIR-24-0084","url":null,"abstract":"<p><p>Epithelial ovarian cancer is the most common type of ovarian cancer with a low rate of response to immunotherapy such as immune checkpoint blockade therapy. In this study, we report that nucleus accumbens-associated protein 1 (NAC1), a putative driver of epithelial ovarian cancer, has a critical role in immune evasion. We showed in murine ovarian cancer models that depleting or inhibiting tumoral NAC1 reduced the recruitment and immunosuppressive function of myeloid-derived suppressor cells (MDSC) in the tumor microenvironment, led to significant increases of cytotoxic tumor-infiltrating CD8+ T cells, and promoted antitumor immunity and suppressed tumor progression. We further showed that tumoral NAC1 directly enhanced the transcription of CXCL16 by binding to CXCR6, thereby promoting MDSC recruitment to the tumor. Moreover, lipid C20:1T produced by NAC1-expressing tumor cells fueled oxidative metabolism of MDSCs and promoted their immune-suppressive function. We also showed that NIC3, a small-molecule inhibitor of NAC1, was able to sensitize mice bearing NAC1-expressing ovarian tumors to anti-PD-1 therapy. Our study reveals a critical role for NAC1 in controlling tumor infiltration of MDSCs and in modulating the efficacy of immune checkpoint blockade therapy. Thus, targeting of NAC1 may be exploited to sensitize ovarian cancer to immunotherapy.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"286-302"},"PeriodicalIF":8.1,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142614540","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
Inflammatory Stress Determines the Need for Chemotherapy in Patients with HER2-Positive Esophagogastric Adenocarcinoma Receiving Targeted Therapy and Immunotherapy. 炎症压力决定了接受靶向和免疫疗法的 HER2 阳性食管胃腺癌患者是否需要化疗。
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2025-02-03 DOI: 10.1158/2326-6066.CIR-24-0561
Joseph Tintelnot, Lisa Paschold, Eray Goekkurt, Christoph Schultheiss, Urte Matschl, Mariana Santos Cruz, Marcus Bauer, Claudia Wickenhauser, Peter Thuss-Patience, Sylvie Lorenzen, Thomas J Ettrich, Jorge Riera-Knorrenschild, Lutz Jacobasch, Albrecht Kretzschmar, Stefan Kubicka, Salah-Eddin Al-Batran, Anke Reinacher-Schick, Daniel Pink, Carsten Bokemeyer, Marianne Sinn, Udo Lindig, Axel Hinke, Susanna Hegewisch-Becker, Alexander Stein, Mascha Binder
{"title":"Inflammatory Stress Determines the Need for Chemotherapy in Patients with HER2-Positive Esophagogastric Adenocarcinoma Receiving Targeted Therapy and Immunotherapy.","authors":"Joseph Tintelnot, Lisa Paschold, Eray Goekkurt, Christoph Schultheiss, Urte Matschl, Mariana Santos Cruz, Marcus Bauer, Claudia Wickenhauser, Peter Thuss-Patience, Sylvie Lorenzen, Thomas J Ettrich, Jorge Riera-Knorrenschild, Lutz Jacobasch, Albrecht Kretzschmar, Stefan Kubicka, Salah-Eddin Al-Batran, Anke Reinacher-Schick, Daniel Pink, Carsten Bokemeyer, Marianne Sinn, Udo Lindig, Axel Hinke, Susanna Hegewisch-Becker, Alexander Stein, Mascha Binder","doi":"10.1158/2326-6066.CIR-24-0561","DOIUrl":"10.1158/2326-6066.CIR-24-0561","url":null,"abstract":"<p><p>Anti-PD-1, trastuzumab, and chemotherapy are used in the treatment of patients with advanced HER2-positive esophagogastric adenocarcinoma, but long-term survival remains limited. In this study, we report extended follow-up data from the INTEGA trial (NCT03409848), which investigated the efficacy of the anti-PD-1 nivolumab, trastuzumab, and FOLFOX chemotherapy (FOLFOX arm) in comparison with a chemotherapy-free regimen involving nivolumab, trastuzumab, and the anti-CTLA-4 ipilimumab (Ipi arm) in the first-line setting for advanced disease. The 12-month overall survival (OS) showed no statistical difference between the arms, with 57% OS (95% confidence interval, 41%-71%) in the Ipi arm and 70% OS (95% confidence interval, 54%-82%) in the FOLFOX arm. Crossing of the survival curves indicated a potential long-term benefit for some patients within the Ipi arm, but early progressors in the Ipi arm underlined the need for biomarker-guided strategies to optimize treatment selection. To this end, metabolomic and cytokine analyses demonstrated elevated levels of normetanephrine, cortisol, and IL6 in immunotherapy-unresponsive patients in the Ipi arm, suggesting a role for systemic inflammatory stress in modulating antitumor immune responses. Patients with this signature also showed an increased neutrophil to lymphocyte ratio that persisted in the Ipi arm, but not in the FOLFOX arm, and strongly correlated with survival. Furthermore, a low neutrophil to lymphocyte ratio characterized patients benefiting from immunotherapy and targeted therapy without the need for additional chemotherapy. These data suggest that patient selection based on inflammatory stress-driven immune changes could help customize first-line treatment in patients with advanced HER2-positive esophagogastric adenocarcinoma to potentially improve long-term survival.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"200-209"},"PeriodicalIF":8.1,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142614424","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.
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2025-02-03 DOI: 10.1158/2326-6066.CIR-13-2-WWR
{"title":"A Sampling of Highlights from the Literature.","authors":"","doi":"10.1158/2326-6066.CIR-13-2-WWR","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-13-2-WWR","url":null,"abstract":"","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":"13 2","pages":"161"},"PeriodicalIF":8.1,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143078750","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
Setdb1 Loss Induces Type I Interferons and Immune Clearance of Melanoma. Setdb1-loss可诱导I型干扰素和黑色素瘤的免疫清除。
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2025-02-03 DOI: 10.1158/2326-6066.CIR-23-0514
Meaghan K McGeary, William Damsky, Andrew J Daniels, Sabine M Lang, Qingji Xu, Eric Song, Clotilde Huet-Calderwood, Hua Jane Lou, Sateja Paradkar, Goran Micevic, Susan M Kaech, David A Calderwood, Benjamin E Turk, Qin Yan, Akiko Iwasaki, Marcus W Bosenberg
{"title":"Setdb1 Loss Induces Type I Interferons and Immune Clearance of Melanoma.","authors":"Meaghan K McGeary, William Damsky, Andrew J Daniels, Sabine M Lang, Qingji Xu, Eric Song, Clotilde Huet-Calderwood, Hua Jane Lou, Sateja Paradkar, Goran Micevic, Susan M Kaech, David A Calderwood, Benjamin E Turk, Qin Yan, Akiko Iwasaki, Marcus W Bosenberg","doi":"10.1158/2326-6066.CIR-23-0514","DOIUrl":"10.1158/2326-6066.CIR-23-0514","url":null,"abstract":"<p><p>Despite recent advances in the treatment of melanoma, many patients with metastatic disease still succumb to their disease. To identify tumor-intrinsic modulators of immunity to melanoma, we performed a whole-genome CRISPR screen in melanoma and identified Setdb1 as well as all components of the human silencing hub complex. We found that loss of Setdb1 leads to increased immunogenicity and complete tumor clearance in a CD8+ T cell-dependent manner. Mechanistically, loss of Setdb1 causes de-repression of endogenous retroviruses (ERV) in melanoma cells and triggers tumor cell-intrinsic type I IFN signaling, upregulation of MHC-I expression, and increased CD8+ T-cell infiltration. Importantly, spontaneous immune clearance observed in Setdb1-/- tumors results in subsequent protection from other ERV-expressing tumor lines, supporting the functional antitumor role of ERV-specific CD8+ T cells found in the Setdb1-/- microenvironment. Blocking the type I IFN receptor in mice grafted with Setdb1-/- tumors decreases immunogenicity by decreasing MHC-I expression, leading to decreased T-cell infiltration and increased melanoma growth, comparable with Setdb1wt tumors. Together, these results provide key in vivo evidence of a critical role for Setdb1 and type I IFNs in generating an inflamed tumor microenvironment and potentiating tumor cell-intrinsic immunogenicity in melanoma. This study further emphasizes regulators of ERV expression and type I IFN expression as potential therapeutic targets for augmenting anticancer immune responses.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"245-257"},"PeriodicalIF":8.1,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142715480","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
Combination CXCR4 and PD-1 Blockade Enhances Intratumoral Dendritic Cell Activation and Immune Responses Against Hepatocellular Carcinoma. CXCR4和PD1联合阻断可增强瘤内树突状细胞活化和对肝细胞癌的免疫反应。
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2025-02-03 DOI: 10.1158/2326-6066.CIR-24-0324
Satoru Morita, Pin-Ji Lei, Kohei Shigeta, Tomofumi Ando, Tatsuya Kobayashi, Hiroto Kikuchi, Aya Matsui, Peigen Huang, Mikael J Pittet, Dan G Duda
{"title":"Combination CXCR4 and PD-1 Blockade Enhances Intratumoral Dendritic Cell Activation and Immune Responses Against Hepatocellular Carcinoma.","authors":"Satoru Morita, Pin-Ji Lei, Kohei Shigeta, Tomofumi Ando, Tatsuya Kobayashi, Hiroto Kikuchi, Aya Matsui, Peigen Huang, Mikael J Pittet, Dan G Duda","doi":"10.1158/2326-6066.CIR-24-0324","DOIUrl":"10.1158/2326-6066.CIR-24-0324","url":null,"abstract":"<p><p>Immune checkpoint inhibitors have revolutionized the treatment of unresectable hepatocellular carcinoma (HCC), but their impressive efficacy is seen in just a fraction of patients. One key mechanism of immunotherapy resistance is the paucity of dendritic cells (DC) in liver malignancies. In this study, we tested combination blockade of PD-1 and CXCR4, a receptor for CXCL12, a pleiotropic factor that mediates immunosuppression in tumors. Using orthotopic grafted and autochthonous HCC models with underlying liver damage, we evaluated treatment feasibility and efficacy. In addition, we examined the effects of treatment using immunofluorescence, flow cytometric analysis of DCs in vivo and in vitro, and RNA sequencing. The combination anti-CXCR4 and anti-PD-1 therapy was safe and significantly inhibited tumor growth and prolonged survival in all murine preclinical models of HCC tested. The combination treatment successfully reprogrammed antigen-presenting cells, revealing the potential role of conventional type 1 DCs (cDC1) in the HCC microenvironment. Moreover, DC reprogramming enhanced anticancer immunity by facilitating CD8+ T-cell accumulation and activation in the HCC tissue. The effectiveness of anti-CXCR4/PD-1 therapy was compromised entirely in Batf3 knockout mice deficient in cDC1s. Thus, combined CXCR4/PD-1 blockade can reprogram intratumoral cDC1s and holds the potential to potentiate antitumor immune response against HCC.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"162-170"},"PeriodicalIF":8.1,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142603039","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 Cells Instruct Immune Checkpoint Inhibitor Therapy Resistance in Tumors Responsive to IL1 and TNFα Inflammation. 肿瘤对IL-1和TNFα炎症反应的免疫检查点抑制剂治疗耐受性的T细胞调控
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2025-02-03 DOI: 10.1158/2326-6066.CIR-24-0416
Nam Woo Cho, Sophia M Guldberg, Barzin Y Nabet, Jie Zeng Yu, Eun Ji Kim, Kamir J Hiam-Galvez, Jacqueline L Yee, Rachel DeBarge, Iliana Tenvooren, Naa Asheley Ashitey, Filipa Lynce, Deborah A Dillon, Jennifer M Rosenbluth, Matthew H Spitzer
{"title":"T Cells Instruct Immune Checkpoint Inhibitor Therapy Resistance in Tumors Responsive to IL1 and TNFα Inflammation.","authors":"Nam Woo Cho, Sophia M Guldberg, Barzin Y Nabet, Jie Zeng Yu, Eun Ji Kim, Kamir J Hiam-Galvez, Jacqueline L Yee, Rachel DeBarge, Iliana Tenvooren, Naa Asheley Ashitey, Filipa Lynce, Deborah A Dillon, Jennifer M Rosenbluth, Matthew H Spitzer","doi":"10.1158/2326-6066.CIR-24-0416","DOIUrl":"10.1158/2326-6066.CIR-24-0416","url":null,"abstract":"<p><p>Resistance to immune checkpoint inhibitors (ICI) is common, even in tumors with T-cell infiltration. We thus investigated consequences of ICI-induced T-cell infiltration in the microenvironment of resistant tumors. T cells and neutrophil numbers increased in ICI-resistant tumors following treatment, in contrast to ICI-responsive tumors. Resistant tumors were distinguished by high expression of IL1 receptor 1, enabling a synergistic response to IL1 and TNFα to induce G-CSF, CXCL1, and CXCL2 via NF-κB signaling, supporting immunosuppressive neutrophil accumulation in tumor. Perturbation of this inflammatory resistance circuit sensitized tumors to ICIs. Paradoxically, T cells drove this resistance circuit via TNFα both in vitro and in vivo. Evidence of this inflammatory resistance circuit and its impact also translated to human cancers. These data support a mechanism of ICI resistance, wherein treatment-induced T-cell activity can drive resistance in tumors responsive to IL1 and TNFα, with important therapeutic implications.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"229-244"},"PeriodicalIF":8.1,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142458701","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信