Pub Date : 2025-09-02DOI: 10.1158/2326-6066.CIR-24-0091
Melinda A Biernacki, Jessica Lok, Kimberly A Foster, Carrie Cummings, Stephanie Busch, R Graeme Black, Suhita Ray, Laura Baquero Galvis, Tim Monahan, Stephen T Oh, Vivian G Oehler, Derek L Stirewalt, David Wu, H Joachim Deeg, Sergei Doulatov, Marie Bleakley
Acquired mutations in spliceosome genes in early hematopoietic stem/progenitor cells are common events in myelodysplastic neoplasms (MDS) and related myeloid malignancies. Mutations in the spliceosome factor subunit B1 (SF3B1) gene occur in ≥20% of MDS cases at conserved hotspots and in early neoplastic clones as driver events. Neoantigens from aberrant SF3B1 proteins could serve as shared T-cell therapy targets for SF3B1-mutated myeloid neoplasms. We identified a candidate neoantigen from the prevalent SF3B1K700E variant using in silico predictions of epitope processing and presentation and then validated presentation and immunogenicity in vitro. CD8+ T cells recognizing SF3B1K700E demonstrated high functional avidity and killed neoplastic myeloid cell lines and primary cells in an antigen-specific manner. We then sequenced, cloned, and transduced an SF3B1K700E-specific T-cell receptor into third-party T cells and confirmed that T-cell receptor transfer conferred antigen specificity and killing of neoplastic myeloid cells in vitro and in vivo. The data indicate that the SF3B1K700E neoantigen represents a promising T-cell target for patients with SF3B1-mutated MDS and acute myeloid leukemia.
{"title":"SF3B1K700E Neoantigen Is a CD8+ T-cell Target Shared across Human Myeloid Neoplasms.","authors":"Melinda A Biernacki, Jessica Lok, Kimberly A Foster, Carrie Cummings, Stephanie Busch, R Graeme Black, Suhita Ray, Laura Baquero Galvis, Tim Monahan, Stephen T Oh, Vivian G Oehler, Derek L Stirewalt, David Wu, H Joachim Deeg, Sergei Doulatov, Marie Bleakley","doi":"10.1158/2326-6066.CIR-24-0091","DOIUrl":"10.1158/2326-6066.CIR-24-0091","url":null,"abstract":"<p><p>Acquired mutations in spliceosome genes in early hematopoietic stem/progenitor cells are common events in myelodysplastic neoplasms (MDS) and related myeloid malignancies. Mutations in the spliceosome factor subunit B1 (SF3B1) gene occur in ≥20% of MDS cases at conserved hotspots and in early neoplastic clones as driver events. Neoantigens from aberrant SF3B1 proteins could serve as shared T-cell therapy targets for SF3B1-mutated myeloid neoplasms. We identified a candidate neoantigen from the prevalent SF3B1K700E variant using in silico predictions of epitope processing and presentation and then validated presentation and immunogenicity in vitro. CD8+ T cells recognizing SF3B1K700E demonstrated high functional avidity and killed neoplastic myeloid cell lines and primary cells in an antigen-specific manner. We then sequenced, cloned, and transduced an SF3B1K700E-specific T-cell receptor into third-party T cells and confirmed that T-cell receptor transfer conferred antigen specificity and killing of neoplastic myeloid cells in vitro and in vivo. The data indicate that the SF3B1K700E neoantigen represents a promising T-cell target for patients with SF3B1-mutated MDS and acute myeloid leukemia.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1391-1404"},"PeriodicalIF":8.2,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144494729","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}
Pub Date : 2025-09-02DOI: 10.1158/2326-6066.CIR-24-1109
Maria T Søgaard, Diane Tseng, Sarah Gibbs, Wei Wu, Lorna G Nolan, Pamela Y Yang, Mason Lai, Jianhong Cao, Sudhakar Pipavath, Koshlan Mayer-Blackwell, Evan W Newell, A McGarry Houghton, Kyle K Payne, Shin-Heng Chiou, Viswam S Nair
The blood T-cell receptor (TCR) repertoire broadly reflects current and lifetime immune responses against infectious pathogens and cancer, but the circulating T-cell repertoire remains a largely untapped resource for cancer biomarker studies due to repertoire complexity and limited profiling data. In this study, we investigated the use of blood TCR sequencing for the early detection of lung cancer. We sequenced the leukocyte fraction of peripheral blood from 633 individuals divided into a case-control cohort (n = 511) and a lung cancer screening cohort (n = 122), representing more than 12.6 million unique clonotypes. Based on the TCR repertoires in these individuals, we devised a Tumor Immune Lymphocyte Score (TILS) using either TCR specificity groups (TILS-A) or highly recurrent "public" TCR clonotypes (TILS-B) capable of detecting lung cancer. TILS-A consisted of 125 TCR specificity groups that outperformed the TILS-B classifier of 49 public, TCRβ-Vβ-defined clonotypes for cancer detection. TILS classifiers (TILS-A and TILS-B) provided predictive value after accounting for age, smoking status, and nodule size in the lung cancer screening cohort and improved cancer prediction for individuals with indeterminate lung cancer risk. In the subgroup analysis, TILS-A was associated with lung cancer in both early- and late-stage disease, had improved accuracy when accounting for HLA status, and was validated in an external dataset studying lung cancer initiation. Collectively, these data suggest that profiles of the circulating T-cell response can provide value for lung cancer detection and support its use as a diagnostic tool.
{"title":"T-cell Receptor Profiling of Blood to Detect Lung Cancer.","authors":"Maria T Søgaard, Diane Tseng, Sarah Gibbs, Wei Wu, Lorna G Nolan, Pamela Y Yang, Mason Lai, Jianhong Cao, Sudhakar Pipavath, Koshlan Mayer-Blackwell, Evan W Newell, A McGarry Houghton, Kyle K Payne, Shin-Heng Chiou, Viswam S Nair","doi":"10.1158/2326-6066.CIR-24-1109","DOIUrl":"10.1158/2326-6066.CIR-24-1109","url":null,"abstract":"<p><p>The blood T-cell receptor (TCR) repertoire broadly reflects current and lifetime immune responses against infectious pathogens and cancer, but the circulating T-cell repertoire remains a largely untapped resource for cancer biomarker studies due to repertoire complexity and limited profiling data. In this study, we investigated the use of blood TCR sequencing for the early detection of lung cancer. We sequenced the leukocyte fraction of peripheral blood from 633 individuals divided into a case-control cohort (n = 511) and a lung cancer screening cohort (n = 122), representing more than 12.6 million unique clonotypes. Based on the TCR repertoires in these individuals, we devised a Tumor Immune Lymphocyte Score (TILS) using either TCR specificity groups (TILS-A) or highly recurrent \"public\" TCR clonotypes (TILS-B) capable of detecting lung cancer. TILS-A consisted of 125 TCR specificity groups that outperformed the TILS-B classifier of 49 public, TCRβ-Vβ-defined clonotypes for cancer detection. TILS classifiers (TILS-A and TILS-B) provided predictive value after accounting for age, smoking status, and nodule size in the lung cancer screening cohort and improved cancer prediction for individuals with indeterminate lung cancer risk. In the subgroup analysis, TILS-A was associated with lung cancer in both early- and late-stage disease, had improved accuracy when accounting for HLA status, and was validated in an external dataset studying lung cancer initiation. Collectively, these data suggest that profiles of the circulating T-cell response can provide value for lung cancer detection and support its use as a diagnostic tool.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1405-1417"},"PeriodicalIF":8.2,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12340770/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144539070","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}
Pub Date : 2025-09-02DOI: 10.1158/2326-6066.CIR-24-1001
Xiaoyu Liu, Xin Liu, Peng Jiang, Xin Qi, Xiuwei Ma, Xiaomin Zhang, Christopher G Proud, Jing Li
NK cells are increasingly being evaluated for their utility in cancer immunotherapy. However, their efficacy is often attenuated in the cancer microenvironment. The identification of additional checkpoint molecules that limit NK cell function is crucial to further development of NK cell-based therapies. In this study, we discovered eukaryotic elongation factor-2 kinase as an important participant in modulating the functional fate of NK cells. Dysfunctional NK cells from patients and tumor-bearing mice were found to have elevated EEF2K expression. CRISPR/Cas9-mediated EEF2K knockout promoted NK cell maturation, proliferation, and cytotoxicity and attenuated their exhaustion. Mechanistic studies demonstrated that EEF2K deletion activated Nrf2 in NK cells, thereby initiating cellular antioxidant signaling to sustain mitochondrial fitness and active metabolism, which was confirmed through combined proteomic high-throughput analysis and experimental observation. In particular, high levels of TGFβ in the tumor microenvironment were found to exacerbate oxidative stress and immunosuppression by inducing EEF2K. Therapeutically, systemic Eef2k deficiency effectively repressed melanoma metastasis and growth while modulating the intratumoral immune microenvironment, and adoptive therapy with EEF2K-knockout NK92 cells exhibited a significant antitumor effect and improved prognosis of human hepatocellular carcinoma xenografts in nude mice. Our findings reveal that eukaryotic elongation factor-2 kinase is an intracellular immune checkpoint of NK cells and provides a potential therapeutic target for developing NK cell-based cancer immunotherapies.
{"title":"Inhibition of eEF-2K Enhances the Antitumor Efficacy of NK Cells.","authors":"Xiaoyu Liu, Xin Liu, Peng Jiang, Xin Qi, Xiuwei Ma, Xiaomin Zhang, Christopher G Proud, Jing Li","doi":"10.1158/2326-6066.CIR-24-1001","DOIUrl":"10.1158/2326-6066.CIR-24-1001","url":null,"abstract":"<p><p>NK cells are increasingly being evaluated for their utility in cancer immunotherapy. However, their efficacy is often attenuated in the cancer microenvironment. The identification of additional checkpoint molecules that limit NK cell function is crucial to further development of NK cell-based therapies. In this study, we discovered eukaryotic elongation factor-2 kinase as an important participant in modulating the functional fate of NK cells. Dysfunctional NK cells from patients and tumor-bearing mice were found to have elevated EEF2K expression. CRISPR/Cas9-mediated EEF2K knockout promoted NK cell maturation, proliferation, and cytotoxicity and attenuated their exhaustion. Mechanistic studies demonstrated that EEF2K deletion activated Nrf2 in NK cells, thereby initiating cellular antioxidant signaling to sustain mitochondrial fitness and active metabolism, which was confirmed through combined proteomic high-throughput analysis and experimental observation. In particular, high levels of TGFβ in the tumor microenvironment were found to exacerbate oxidative stress and immunosuppression by inducing EEF2K. Therapeutically, systemic Eef2k deficiency effectively repressed melanoma metastasis and growth while modulating the intratumoral immune microenvironment, and adoptive therapy with EEF2K-knockout NK92 cells exhibited a significant antitumor effect and improved prognosis of human hepatocellular carcinoma xenografts in nude mice. Our findings reveal that eukaryotic elongation factor-2 kinase is an intracellular immune checkpoint of NK cells and provides a potential therapeutic target for developing NK cell-based cancer immunotherapies.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1485-1501"},"PeriodicalIF":8.2,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144282448","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}
Tyrosine kinase inhibitors are initially efficacious against anaplastic lymphoma kinase (ALK) fusion gene-positive lung adenocarcinoma, but acquired resistance inevitably occurs. Therefore, alternative treatment strategies are needed for tyrosine kinase inhibitor-resistant cases. Although the use of immune checkpoint inhibitors (ICI) has improved the prognosis of patients with lung cancer, patients with ALK+ lung adenocarcinoma exhibit little or no response to immunotherapy and the underlying resistance mechanisms remain unknown. In this study, we explored the immunologic status of the tumor microenvironment (TME) in ALK+ lung adenocarcinoma tissues. Tumor-infiltrating leukocyte analysis revealed reduced numbers of effector T cells and increased myeloid-derived suppressor cells (MDSC) relative to ALK- lung adenocarcinoma cases, indicating that ALK+ lung adenocarcinoma has a myeloid cell-dominant immunosuppressive TME. Single-cell RNA sequencing analysis identified a subset of macrophages that expressed most T cell-attractant chemokines (CXCL9, CXCL10, and CXCL11), and the macrophages were inactivated in ALK+ lung adenocarcinoma. In contrast, ALK+ lung adenocarcinoma expressed high levels of MDSC-attractant chemokines (CXCL1 and CXCL8). In addition, ALK+ lung adenocarcinoma showed higher levels of IL6, an MDSC-inducing cytokine, than ALK- lung adenocarcinoma. An IL6R inhibitor transformed the TME in a murine ALK+ lung adenocarcinoma model, shifting it from an immunosuppressive to a T cell-dominant status. Although ICI monotherapy lacked antitumor effects, a combination of ICI and the IL6R inhibitor had significant antitumor effects in mice. Our findings illustrate the molecular basis of fusion gene-mediated immunosuppressive TMEs, providing a rationale for a novel combination immunotherapy for ALK+ lung adenocarcinoma. See related Spotlight by Vitale and Bria, p.1326.
{"title":"EML4-ALK Rearrangement Creates a Distinctive Myeloid Cell-Dominant Immunosuppressive Microenvironment in Lung Cancer.","authors":"Kosuke Arai, Yukari Nishito, Hideaki Mizuno, Noriko Motoi, Nobuyoshi Hiraoka, Masanori Fuse, Yasuhito Arai, Tatsuhiro Shibata, Yukiko Sonobe, Yoko Kayukawa, Toru Maruyama, Hironori Fukuda, Yukihiro Mizoguchi, Yukiko Aikawa, Yukihiro Yoshida, Shun-Ichi Watanabe, Hiromi Sakamoto, Makiko Yamashita, Shigehisa Kitano, Yuki Nagata, Risa Mitsumori, Kouichi Ozaki, Shumpei Niida, Yae Kanai, Akiyoshi Hirayama, Tomoyoshi Soga, Teruhiko Yoshida, Kazuki Yasuda, Atsushi Ochiai, Hiroyuki Tsunoda, Kazunori Aoki","doi":"10.1158/2326-6066.CIR-24-0532","DOIUrl":"10.1158/2326-6066.CIR-24-0532","url":null,"abstract":"<p><p>Tyrosine kinase inhibitors are initially efficacious against anaplastic lymphoma kinase (ALK) fusion gene-positive lung adenocarcinoma, but acquired resistance inevitably occurs. Therefore, alternative treatment strategies are needed for tyrosine kinase inhibitor-resistant cases. Although the use of immune checkpoint inhibitors (ICI) has improved the prognosis of patients with lung cancer, patients with ALK+ lung adenocarcinoma exhibit little or no response to immunotherapy and the underlying resistance mechanisms remain unknown. In this study, we explored the immunologic status of the tumor microenvironment (TME) in ALK+ lung adenocarcinoma tissues. Tumor-infiltrating leukocyte analysis revealed reduced numbers of effector T cells and increased myeloid-derived suppressor cells (MDSC) relative to ALK- lung adenocarcinoma cases, indicating that ALK+ lung adenocarcinoma has a myeloid cell-dominant immunosuppressive TME. Single-cell RNA sequencing analysis identified a subset of macrophages that expressed most T cell-attractant chemokines (CXCL9, CXCL10, and CXCL11), and the macrophages were inactivated in ALK+ lung adenocarcinoma. In contrast, ALK+ lung adenocarcinoma expressed high levels of MDSC-attractant chemokines (CXCL1 and CXCL8). In addition, ALK+ lung adenocarcinoma showed higher levels of IL6, an MDSC-inducing cytokine, than ALK- lung adenocarcinoma. An IL6R inhibitor transformed the TME in a murine ALK+ lung adenocarcinoma model, shifting it from an immunosuppressive to a T cell-dominant status. Although ICI monotherapy lacked antitumor effects, a combination of ICI and the IL6R inhibitor had significant antitumor effects in mice. Our findings illustrate the molecular basis of fusion gene-mediated immunosuppressive TMEs, providing a rationale for a novel combination immunotherapy for ALK+ lung adenocarcinoma. See related Spotlight by Vitale and Bria, p.1326.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1435-1452"},"PeriodicalIF":8.2,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144590507","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}
Pub Date : 2025-09-02DOI: 10.1158/2326-6066.CIR-25-0624
Antonio Vitale, Emilio Bria
Anaplastic lymphoma kinase-rearranged lung adenocarcinoma (ALK+ LUAD) is currently considered an immune-resistant disease, yet underlying biological mechanisms are largely unknown. In this issue, Arai and colleagues analyzed the tumor microenvironment (TME) in ALK+ LUADs, identifying a myeloid cell-dominant immunosuppressive TME, primarily driven by IL6 secretion. Dual anti-IL6R/anti-PD-L1 treatment resulted in robust antitumor effect in mouse models, restoring immune sensitivity and tumor control. These findings highlight a promising therapeutic approach to enhance the efficacy of PD-(L)1 inhibitors by reverting TME-mediated immune resistance, reshaping the role of immunotherapy in ALK+ LUADs. See related article by Arai et al., p. 1435.
{"title":"Unlocking the Immune Response in ALK-Rearranged Lung Adenocarcinoma.","authors":"Antonio Vitale, Emilio Bria","doi":"10.1158/2326-6066.CIR-25-0624","DOIUrl":"10.1158/2326-6066.CIR-25-0624","url":null,"abstract":"<p><p>Anaplastic lymphoma kinase-rearranged lung adenocarcinoma (ALK+ LUAD) is currently considered an immune-resistant disease, yet underlying biological mechanisms are largely unknown. In this issue, Arai and colleagues analyzed the tumor microenvironment (TME) in ALK+ LUADs, identifying a myeloid cell-dominant immunosuppressive TME, primarily driven by IL6 secretion. Dual anti-IL6R/anti-PD-L1 treatment resulted in robust antitumor effect in mouse models, restoring immune sensitivity and tumor control. These findings highlight a promising therapeutic approach to enhance the efficacy of PD-(L)1 inhibitors by reverting TME-mediated immune resistance, reshaping the role of immunotherapy in ALK+ LUADs. See related article by Arai et al., p. 1435.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1326-1327"},"PeriodicalIF":8.2,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144798235","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}
Pub Date : 2025-09-02DOI: 10.1158/2326-6066.CIR-13-9-WWR
{"title":"A Sampling of Highlights from the Literature: Article Recommendations from Our Deputy and Senior Editors.","authors":"","doi":"10.1158/2326-6066.CIR-13-9-WWR","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-13-9-WWR","url":null,"abstract":"","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":"13 9","pages":"1325"},"PeriodicalIF":8.2,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144944040","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}
Cancer-associated fibroblasts (CAF) play immunosuppressive roles in the tumor microenvironment. Specifically, they reportedly act as physical barriers preventing immune cell infiltration. However, the spatial relationships between CAFs and cancer cells in antitumor immunity remain unknown. In this study, we established three-dimensional (3D) constructs, in which the spatial relationships were controlled using a 3D bioprinter. Using these models, we found that the mixed distribution of fibroblasts (FB) and cancer cells suppressed the antitumor immunity more than the surrounding distribution of FBs as physical barriers. The 3D construct with mixed distribution promoted TGFβ and periostin (encoded by Postn gene) cross-talk, resulting in immunosuppression. Postn knockdown in FBs decreased the TGFβ production in the mixed 3D construct and activated antitumor immunity both in vitro and in vivo. Clinically, patients with head and neck cancer or lung cancer showing a mixed distribution of α-smooth muscle actin+ myofibroblast-like CAFs exhibited worse prognosis after PD-1 blockade therapies, and lower CD8+ T-cell infiltration than those that had CAFs surrounding cancer cells. Overall, our findings suggest that the close interactions of CAFs and cancer cells facilitate immunosuppression, rather than the physical barriers created by CAFs, highlighting their potential as biomarkers and therapeutic targets for cancer immunotherapies based on spatial relationships. Furthermore, this study highlights the beneficial applications of 3D bioprinters.
{"title":"Close Spatial Interactions between Cancer Cells and Cancer-Associated Fibroblasts Suppress Antitumor Immunity.","authors":"Yuto Naoi, Yumi Inukai, Tomoka Izumikawa, Joji Nagasaki, Takamasa Ishino, Youki Ueda, Yin Min Thu, Miho Fujiwara, Takahiro Baba, Go Makimoto, Ken Suzawa, Kazuhiro Okada, Ken-Ichi Yamamoto, Masakiyo Sakaguchi, Shuta Tomida, Yoshinobu Maeda, Shinichi Toyooka, Mizuo Ando, Yosuke Togashi","doi":"10.1158/2326-6066.CIR-24-1144","DOIUrl":"10.1158/2326-6066.CIR-24-1144","url":null,"abstract":"<p><p>Cancer-associated fibroblasts (CAF) play immunosuppressive roles in the tumor microenvironment. Specifically, they reportedly act as physical barriers preventing immune cell infiltration. However, the spatial relationships between CAFs and cancer cells in antitumor immunity remain unknown. In this study, we established three-dimensional (3D) constructs, in which the spatial relationships were controlled using a 3D bioprinter. Using these models, we found that the mixed distribution of fibroblasts (FB) and cancer cells suppressed the antitumor immunity more than the surrounding distribution of FBs as physical barriers. The 3D construct with mixed distribution promoted TGFβ and periostin (encoded by Postn gene) cross-talk, resulting in immunosuppression. Postn knockdown in FBs decreased the TGFβ production in the mixed 3D construct and activated antitumor immunity both in vitro and in vivo. Clinically, patients with head and neck cancer or lung cancer showing a mixed distribution of α-smooth muscle actin+ myofibroblast-like CAFs exhibited worse prognosis after PD-1 blockade therapies, and lower CD8+ T-cell infiltration than those that had CAFs surrounding cancer cells. Overall, our findings suggest that the close interactions of CAFs and cancer cells facilitate immunosuppression, rather than the physical barriers created by CAFs, highlighting their potential as biomarkers and therapeutic targets for cancer immunotherapies based on spatial relationships. Furthermore, this study highlights the beneficial applications of 3D bioprinters.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1471-1484"},"PeriodicalIF":8.2,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144539068","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}
Pub Date : 2025-09-02DOI: 10.1158/2326-6066.CIR-24-1128
Miki Yamada-Hara, Lauren Amaya, Zhihe Wang, Ji Won Byun, Naoki Takahashi, Sunandini Sharma, Han Chang, Arisachi Tanaka, Liping Zeng, Zahra Malakoutikhah, Sneha Ganguly, Minh-Chau Vu, Matt Levin, David Schwartz, Jack Heath, Scott Herdman, Maripat Corr, Eyal Raz, Samuel Bertin
The retinoic acid receptor-related orphan receptor C (RORC) gene encodes two isoforms, RORγ and RORγt, which function as transcription factors in different cell types. RORγt is expressed in specific immune cells involved in inflammatory responses, whereas RORγ is found in parenchymal cells, in which it participates in metabolism and circadian rhythm regulation. Although the roles of RORγt in CD4+ Th17 lymphocytes and RORγ in certain cancer cell types are increasingly recognized, their relative contributions to lung cancer development remain unclear. In this study, we investigated the roles of RORC, RORγ, and RORγt in lung cancer using mouse models and human data from The Cancer Genome Atlas. We evaluated the effects of Rorc gene deletion and RORγ/γt pharmacologic inhibition in cancer and immune cells in vitro and in vivo. Pharmacologic blockade of RORγ/γt with digoxin significantly reduced lung cancer development in two mouse models: a KrasG12D-driven genetic model and a urethane-induced chemical model. Mechanistically, this effect was mediated by inhibition of RORγt in specific immune cells, such as type 3 innate lymphoid cells and Th17 cells, rather than by inhibiting RORγ in tumor cells. This reduced the production of proinflammatory cytokines, including IL17A, IL17F, and IL22, and decreased tumor cell proliferation. Additionally, The Cancer Genome Atlas analysis revealed that elevated RORC expression is associated with an altered tumor microenvironment and poorer prognosis in patients with lung adenocarcinoma. These findings highlight the therapeutic potential of targeting RORγt to reduce protumor inflammation and propose a strategy for lung cancer treatment.
视黄酸受体相关孤儿受体C (RORC)基因编码RORγ和RORγt两个亚型,在不同细胞类型中发挥转录因子的作用。RORγt在参与炎症反应的特异性免疫细胞中表达,而RORγ在实质细胞中发现,参与代谢和昼夜节律调节。尽管RORγt在CD4+ t -辅助性17 (Th17)淋巴细胞中的作用和RORγ在某些癌症细胞类型中的作用越来越被认识到,但它们在肺癌(LC)发展中的相对作用仍不清楚。在这项研究中,我们利用来自癌症基因组图谱(TCGA)的小鼠模型和人类数据,研究了RORC、RORγ和RORγt在LC中的作用。我们在体外和体内评估了Rorc基因缺失和RORγ/γt药理抑制对肿瘤和免疫细胞的影响。在krasg12d驱动的遗传模型和聚氨酯诱导的化学模型中,地高辛阻断rorr γ/γt可显著降低两种小鼠模型的LC发展。在机制上,这种作用是通过抑制特异性免疫细胞(如3型先天淋巴样细胞(ILC3s)和Th17细胞)中的RORγt而不是抑制肿瘤细胞中的RORγ来介导的。这减少了促炎细胞因子的产生,包括白细胞介素- 17a (IL-17A)、IL-17F和IL-22,并降低了肿瘤细胞的增殖。此外,TCGA分析显示,RORC表达升高与肺腺癌(LUAD)患者肿瘤微环境(TME)改变和预后较差有关。这些发现强调了靶向RORγt减少肿瘤前炎症的治疗潜力,并提出了LC治疗的策略。
{"title":"RORγt Inhibition Reduces Protumor Inflammation and Decreases Tumor Growth in Experimental Models of Lung Cancer.","authors":"Miki Yamada-Hara, Lauren Amaya, Zhihe Wang, Ji Won Byun, Naoki Takahashi, Sunandini Sharma, Han Chang, Arisachi Tanaka, Liping Zeng, Zahra Malakoutikhah, Sneha Ganguly, Minh-Chau Vu, Matt Levin, David Schwartz, Jack Heath, Scott Herdman, Maripat Corr, Eyal Raz, Samuel Bertin","doi":"10.1158/2326-6066.CIR-24-1128","DOIUrl":"10.1158/2326-6066.CIR-24-1128","url":null,"abstract":"<p><p>The retinoic acid receptor-related orphan receptor C (RORC) gene encodes two isoforms, RORγ and RORγt, which function as transcription factors in different cell types. RORγt is expressed in specific immune cells involved in inflammatory responses, whereas RORγ is found in parenchymal cells, in which it participates in metabolism and circadian rhythm regulation. Although the roles of RORγt in CD4+ Th17 lymphocytes and RORγ in certain cancer cell types are increasingly recognized, their relative contributions to lung cancer development remain unclear. In this study, we investigated the roles of RORC, RORγ, and RORγt in lung cancer using mouse models and human data from The Cancer Genome Atlas. We evaluated the effects of Rorc gene deletion and RORγ/γt pharmacologic inhibition in cancer and immune cells in vitro and in vivo. Pharmacologic blockade of RORγ/γt with digoxin significantly reduced lung cancer development in two mouse models: a KrasG12D-driven genetic model and a urethane-induced chemical model. Mechanistically, this effect was mediated by inhibition of RORγt in specific immune cells, such as type 3 innate lymphoid cells and Th17 cells, rather than by inhibiting RORγ in tumor cells. This reduced the production of proinflammatory cytokines, including IL17A, IL17F, and IL22, and decreased tumor cell proliferation. Additionally, The Cancer Genome Atlas analysis revealed that elevated RORC expression is associated with an altered tumor microenvironment and poorer prognosis in patients with lung adenocarcinoma. These findings highlight the therapeutic potential of targeting RORγt to reduce protumor inflammation and propose a strategy for lung cancer treatment.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1418-1434"},"PeriodicalIF":8.2,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12288671/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144324574","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}
Pub Date : 2025-09-02DOI: 10.1158/2326-6066.CIR-25-0034
Sreekumar Balan, Liam O'Brien, Ante Peros, Xuedi Wang, Ingrid Leal Rojas, Christopher McClain, Kristen J Radford, Nina Bhardwaj
A dendritic cell (DC)-based vaccine, Sipuleucel-T, remains the sole FDA-approved cancer vaccine. Despite their established safety and efficacy against cancers and infections in numerous trials, long-term clinical benefits have been modest. Most trials have employed DCs derived from blood monocytes, but emerging evidence underscores the unique role of conventional type 1 DCs (cDC1) in triggering potent antitumor immune responses and their intratumoral infiltration with favorable prognoses in many cancers. However, the scarcity of cDC1s in peripheral blood and the challenges in generating them in vitro have hindered a deeper understanding of their biology and their widespread application as cellular vaccines. In this study, we present a serum-free culture system capable of generating billions of human cDC1s from CD34+ progenitors derived from cord or peripheral blood. The system leverages the requirement of Notch signaling for cDC1 differentiation and generates DCs that closely resemble in vivo cDC1s, exhibiting functions including cellular antigen cross-presentation. This robust protocol enables the scalable production of cDC1s for both fundamental biological research and therapeutic applications.
{"title":"Harnessing Notch Signaling to Enhance the Generation and Functionality of Human Conventional Type 1 Dendritic Cells for Cancer Immunotherapy Applications.","authors":"Sreekumar Balan, Liam O'Brien, Ante Peros, Xuedi Wang, Ingrid Leal Rojas, Christopher McClain, Kristen J Radford, Nina Bhardwaj","doi":"10.1158/2326-6066.CIR-25-0034","DOIUrl":"10.1158/2326-6066.CIR-25-0034","url":null,"abstract":"<p><p>A dendritic cell (DC)-based vaccine, Sipuleucel-T, remains the sole FDA-approved cancer vaccine. Despite their established safety and efficacy against cancers and infections in numerous trials, long-term clinical benefits have been modest. Most trials have employed DCs derived from blood monocytes, but emerging evidence underscores the unique role of conventional type 1 DCs (cDC1) in triggering potent antitumor immune responses and their intratumoral infiltration with favorable prognoses in many cancers. However, the scarcity of cDC1s in peripheral blood and the challenges in generating them in vitro have hindered a deeper understanding of their biology and their widespread application as cellular vaccines. In this study, we present a serum-free culture system capable of generating billions of human cDC1s from CD34+ progenitors derived from cord or peripheral blood. The system leverages the requirement of Notch signaling for cDC1 differentiation and generates DCs that closely resemble in vivo cDC1s, exhibiting functions including cellular antigen cross-presentation. This robust protocol enables the scalable production of cDC1s for both fundamental biological research and therapeutic applications.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1328-1341"},"PeriodicalIF":8.2,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144539069","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}
Pub Date : 2025-09-02DOI: 10.1158/2326-6066.CIR-24-0975
Reona Okada, Jeyshka M Reyes-González, Constanza Rodriguez, Taisuke Kondo, Jangsuk Oh, Ming Sun, Michael C Kelly, Ling Zhang, James Gulley, Jack F Shern, Mitchell Ho, Christian S Hinrichs, Naomi N Taylor, Xiyuan Zhang, Rosa Nguyen
Neuroblastoma is a highly aggressive childhood solid tumor with poor outcomes. Chimeric antigen receptor (CAR) T cells have shown limited efficacy in neuroblastoma, with the best outcomes reported in patients with a low tumor burden, highlighting the need for further CAR optimization. One approach to addressing the high tumor burden involves engineering CAR T cells to release or express transgenic cytokines. However, its systemic toxicity remains an important therapeutic challenge. In this study, we evaluated the efficacy of IL15- and IL21-enhanced glypican 2 (GPC2)-targeted CAR T cells (GPC2-CAR T cells) in targeting high-burden neuroblastoma. Three strategies for expressing the cytokines were evaluated: constitutive secretion (GPC2-CAR + sol.IL15.IL21), constitutive membrane-tethered expression (GPC2-CAR + teth.IL15.IL21), and NFAT-inducible membrane-tethered expression (GPC2-CAR + NFAT.IL15.IL21). Engineered GPC2-CAR T cells were tested in vitro and in vivo using high neuroblastoma burden xenograft models. Additionally, single-cell RNA sequencing was used to profile the effector cells in the tumor microenvironment. All three versions of GPC2-CAR T cells significantly enhanced killing against a high neuroblastoma burden, both in vitro and in vivo, relative to control GPC2-CAR T cells. Mice treated with GPC2-CAR + NFAT.IL15.IL21 exhibited significantly lower anorexia-associated morbidity/mortality. Supporting these data, tumor-infiltrating GPC2-CAR + NFAT.IL15.IL21 developed an immunosuppressive transcriptional profile upon tumor regression, leading to prolonged survival in treated mice. In contrast, GPC2-CAR + teth.IL15.IL21 maintained a proinflammatory transcriptional signature despite near tumor clearance, resulting in hypercytokinemia and death. NFAT-inducible co-expression of tethered IL15/IL21 enhanced GPC2-CAR T-cell function against a high neuroblastoma burden with acceptable tolerability in mice. Further studies are required to validate these findings.
{"title":"GPC2-Targeted CAR T Cells Engineered with NFAT-Inducible Membrane-Tethered IL15/IL21 Exhibit Enhanced Activity against Neuroblastoma.","authors":"Reona Okada, Jeyshka M Reyes-González, Constanza Rodriguez, Taisuke Kondo, Jangsuk Oh, Ming Sun, Michael C Kelly, Ling Zhang, James Gulley, Jack F Shern, Mitchell Ho, Christian S Hinrichs, Naomi N Taylor, Xiyuan Zhang, Rosa Nguyen","doi":"10.1158/2326-6066.CIR-24-0975","DOIUrl":"10.1158/2326-6066.CIR-24-0975","url":null,"abstract":"<p><p>Neuroblastoma is a highly aggressive childhood solid tumor with poor outcomes. Chimeric antigen receptor (CAR) T cells have shown limited efficacy in neuroblastoma, with the best outcomes reported in patients with a low tumor burden, highlighting the need for further CAR optimization. One approach to addressing the high tumor burden involves engineering CAR T cells to release or express transgenic cytokines. However, its systemic toxicity remains an important therapeutic challenge. In this study, we evaluated the efficacy of IL15- and IL21-enhanced glypican 2 (GPC2)-targeted CAR T cells (GPC2-CAR T cells) in targeting high-burden neuroblastoma. Three strategies for expressing the cytokines were evaluated: constitutive secretion (GPC2-CAR + sol.IL15.IL21), constitutive membrane-tethered expression (GPC2-CAR + teth.IL15.IL21), and NFAT-inducible membrane-tethered expression (GPC2-CAR + NFAT.IL15.IL21). Engineered GPC2-CAR T cells were tested in vitro and in vivo using high neuroblastoma burden xenograft models. Additionally, single-cell RNA sequencing was used to profile the effector cells in the tumor microenvironment. All three versions of GPC2-CAR T cells significantly enhanced killing against a high neuroblastoma burden, both in vitro and in vivo, relative to control GPC2-CAR T cells. Mice treated with GPC2-CAR + NFAT.IL15.IL21 exhibited significantly lower anorexia-associated morbidity/mortality. Supporting these data, tumor-infiltrating GPC2-CAR + NFAT.IL15.IL21 developed an immunosuppressive transcriptional profile upon tumor regression, leading to prolonged survival in treated mice. In contrast, GPC2-CAR + teth.IL15.IL21 maintained a proinflammatory transcriptional signature despite near tumor clearance, resulting in hypercytokinemia and death. NFAT-inducible co-expression of tethered IL15/IL21 enhanced GPC2-CAR T-cell function against a high neuroblastoma burden with acceptable tolerability in mice. Further studies are required to validate these findings.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"1363-1373"},"PeriodicalIF":8.2,"publicationDate":"2025-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12278764/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144494728","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}