Cancer immunology research最新文献

筛选
英文 中文
The Exonuclease TREX1 Constitutes an Innate Immune Checkpoint Limiting cGAS/STING-Mediated Antitumor Immunity. 外切酶 TREX1 是限制 cGAS/STING 介导的抗肿瘤免疫的先天免疫检查点。
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-23-1078
Junghyun Lim, Ryan Rodriguez, Katherine Williams, John Silva, Alan G Gutierrez, Paul Tyler, Faezzah Baharom, Tao Sun, Eva Lin, Scott Martin, Brandon D Kayser, Robert J Johnston, Ira Mellman, Lélia Delamarre, Nathaniel R West, Sören Müller, Yan Qu, Klaus Heger
{"title":"The Exonuclease TREX1 Constitutes an Innate Immune Checkpoint Limiting cGAS/STING-Mediated Antitumor Immunity.","authors":"Junghyun Lim, Ryan Rodriguez, Katherine Williams, John Silva, Alan G Gutierrez, Paul Tyler, Faezzah Baharom, Tao Sun, Eva Lin, Scott Martin, Brandon D Kayser, Robert J Johnston, Ira Mellman, Lélia Delamarre, Nathaniel R West, Sören Müller, Yan Qu, Klaus Heger","doi":"10.1158/2326-6066.CIR-23-1078","DOIUrl":"10.1158/2326-6066.CIR-23-1078","url":null,"abstract":"<p><p>The DNA exonuclease three-prime repair exonuclease 1 (TREX1) is critical for preventing autoimmunity in mice and humans by degrading endogenous cytosolic DNA, which otherwise triggers activation of the innate cGAS/STING pathway leading to the production of type I IFNs. As tumor cells are prone to aberrant cytosolic DNA accumulation, we hypothesized that they are critically dependent on TREX1 activity to limit their immunogenicity. Here, we show that in tumor cells, TREX1 restricts spontaneous activation of the cGAS/STING pathway, and the subsequent induction of a type I IFN response. As a result, TREX1 deficiency compromised in vivo tumor growth in mice. This delay in tumor growth depended on a functional immune system, systemic type I IFN signaling, and tumor-intrinsic cGAS expression. Mechanistically, we show that tumor TREX1 loss drove activation of CD8+ T cells and NK cells, prevented CD8+ T-cell exhaustion, and remodeled an immunosuppressive myeloid compartment. Consequently, TREX1 deficiency combined with T-cell-directed immune checkpoint blockade. Collectively, we conclude that TREX1 is essential to limit tumor immunogenicity, and that targeting this innate immune checkpoint remodels the tumor microenvironment and enhances antitumor immunity by itself and in combination with T-cell-targeted therapies. See related article by Toufektchan et al., p. 673.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"663-672"},"PeriodicalIF":8.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11148535/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140136498","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The ω-3 Polyunsaturated Fatty Acid Docosahexaenoic Acid Enhances NK-Cell Antitumor Effector Functions. ω-3多不饱和脂肪酸二十二碳六烯酸能增强NK细胞的抗肿瘤效应功能。
IF 10.1 1区 医学
Cancer immunology research Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-23-0359
Shuting Wu, Hongyan Peng, Songyang Li, Lanlan Huang, Xiangyu Wang, Yana Li, Yongjie Liu, Peiwen Xiong, Qinglan Yang, Kunpeng Tian, Weiru Wu, Rongxi Pu, Xiulan Lu, Zhenghui Xiao, Jian Yang, Zhaoyang Zhong, Yuan Gao, Yafei Deng, Youcai Deng
{"title":"The ω-3 Polyunsaturated Fatty Acid Docosahexaenoic Acid Enhances NK-Cell Antitumor Effector Functions.","authors":"Shuting Wu, Hongyan Peng, Songyang Li, Lanlan Huang, Xiangyu Wang, Yana Li, Yongjie Liu, Peiwen Xiong, Qinglan Yang, Kunpeng Tian, Weiru Wu, Rongxi Pu, Xiulan Lu, Zhenghui Xiao, Jian Yang, Zhaoyang Zhong, Yuan Gao, Yafei Deng, Youcai Deng","doi":"10.1158/2326-6066.CIR-23-0359","DOIUrl":"10.1158/2326-6066.CIR-23-0359","url":null,"abstract":"<p><p>ω-3 polyunsaturated fatty acids (PUFA) are known to directly repress tumor development and progression. In this study, we explored whether docosahexaenoic acid (DHA), a type of ω-3 PUFA, had an immunomodulatory role in inhibiting tumor growth in immunocompetent mice. The number of natural killer (NK) cells but not the number of T or B cells was decreased by DHA supplementation in various tissues under physiologic conditions. Although the frequency and number of NK cells were comparable, IFNγ production by NK cells in both the spleen and lung was increased in DHA-supplemented mice in the mouse B16F10 melanoma tumor model. Single-cell RNA sequencing revealed that DHA promoted effector function and oxidative phosphorylation in NK cells but had no obvious effects on other immune cells. Using Rag2-/- mice and NK-cell depletion by PK136 antibody injection, we demonstrated that the suppression of B16F10 melanoma tumor growth in the lung by DHA supplementation was dependent mainly on NK cells. In vitro experiments showed that DHA directly enhanced IFNγ production, CD107a expression, and mitochondrial oxidative phosphorylation (OXPHOS) activity and slightly increased proliferator-activated receptor gamma coactivator-1α (PGC-1α) protein expression in NK cells. The PGC-1α inhibitor SR-18292 in vitro and NK cell-specific knockout of PGC-1α in mice reversed the antitumor effects of DHA. In summary, our findings broaden the current knowledge on how DHA supplementation protects against cancer growth from the perspective of immunomodulation by upregulating PGC-1α signaling-mediated mitochondrial OXPHOS activity in NK cells.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"744-758"},"PeriodicalIF":10.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11148550/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140206337","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Intratumoral TREX1 Induction Promotes Immune Evasion by Limiting Type I IFN. 瘤内TREX1诱导通过限制I型干扰素促进免疫逃避。
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-23-1093
Eléonore Toufektchan, Alexandra Dananberg, Josefine Striepen, James H Hickling, Abraham Shim, Yanyang Chen, Ashley Nichols, Mercedes A Duran Paez, Lisa Mohr, Samuel F Bakhoum, John Maciejowski
{"title":"Intratumoral TREX1 Induction Promotes Immune Evasion by Limiting Type I IFN.","authors":"Eléonore Toufektchan, Alexandra Dananberg, Josefine Striepen, James H Hickling, Abraham Shim, Yanyang Chen, Ashley Nichols, Mercedes A Duran Paez, Lisa Mohr, Samuel F Bakhoum, John Maciejowski","doi":"10.1158/2326-6066.CIR-23-1093","DOIUrl":"10.1158/2326-6066.CIR-23-1093","url":null,"abstract":"<p><p>Chromosomal instability is a hallmark of human cancer that is associated with aggressive disease characteristics. Chromosome mis-segregations help fuel natural selection, but they risk provoking a cGAS-STING immune response through the accumulation of cytosolic DNA. The mechanisms of how tumors benefit from chromosomal instability while mitigating associated risks, such as enhanced immune surveillance, are poorly understood. Here, we identify cGAS-STING-dependent upregulation of the nuclease TREX1 as an adaptive, negative feedback mechanism that promotes immune evasion through digestion of cytosolic DNA. TREX1 loss diminishes tumor growth, prolongs survival of host animals, increases tumor immune infiltration, and potentiates response to immune checkpoint blockade selectively in tumors capable of mounting a type I IFN response downstream of STING. Together, these data demonstrate that TREX1 induction shields chromosomally unstable tumors from immune surveillance by dampening type I IFN production and suggest that TREX1 inhibitors might be used to selectively target tumors that have retained the inherent ability to mount an IFN response downstream of STING. See related article by Lim et al., p. 663.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"673-686"},"PeriodicalIF":8.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11148545/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139971036","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Randomized Phase II Study Evaluating the Addition of Pembrolizumab to Radium-223 in Metastatic Castration-resistant Prostate Cancer. 评估转移性钙化抗性前列腺癌患者在镭-223基础上加用 Pembrolizumab 的随机 II 期研究。
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-22-0306
Atish D Choudhury, Lucia Kwak, Alexander Cheung, Kathryn M Allaire, Jaqueline Marquez, David D Yang, Abhishek Tripathi, Jacqueline M Kilar, Meredith Flynn, Brianna Maynard, Rebecca Reichel, Amanda F Pace, Brandon K Chen, Eliezer M Van Allen, Kerry Kilbridge, Xiao X Wei, Bradley A McGregor, Mark M Pomerantz, Rupal S Bhatt, Christopher J Sweeney, Glenn J Bubley, Heather A Jacene, Mary-Ellen Taplin, Franklin W Huang, Lauren C Harshman, Lawrence Fong
{"title":"Randomized Phase II Study Evaluating the Addition of Pembrolizumab to Radium-223 in Metastatic Castration-resistant Prostate Cancer.","authors":"Atish D Choudhury, Lucia Kwak, Alexander Cheung, Kathryn M Allaire, Jaqueline Marquez, David D Yang, Abhishek Tripathi, Jacqueline M Kilar, Meredith Flynn, Brianna Maynard, Rebecca Reichel, Amanda F Pace, Brandon K Chen, Eliezer M Van Allen, Kerry Kilbridge, Xiao X Wei, Bradley A McGregor, Mark M Pomerantz, Rupal S Bhatt, Christopher J Sweeney, Glenn J Bubley, Heather A Jacene, Mary-Ellen Taplin, Franklin W Huang, Lauren C Harshman, Lawrence Fong","doi":"10.1158/2326-6066.CIR-22-0306","DOIUrl":"10.1158/2326-6066.CIR-22-0306","url":null,"abstract":"<p><p>The checkpoint immunotherapeutic pembrolizumab induces responses in a small minority of patients with metastatic castration-resistant prostate cancer (mCRPC). Radium-223 (R223) may increase immunogenicity of bone metastases and increase pembrolizumab (P) activity. In a randomized phase II study, we assessed the effect of R223+P compared with R223 on tumor immune infiltration, safety, and clinical outcomes in patients with mCRPC. The primary endpoint was differences in CD4+ and CD8+ T-cell infiltrate in 8-week versus baseline bone metastasis biopsies; secondary endpoints were safety, radiographic progression-free survival (rPFS), and overall survival (OS). Of the 42 treated patients (29 R223+P, 13 R223), 18 R223+P and 8 R223 patients had evaluable paired tumor biopsies. Median fold-change of CD4+ T cells was -0.7 (range: -9.3 to 4.7) with R223+P and 0.1 (-11.1 to 3.7) with R223 (P = 0.66); for CD8+ T cells, median fold-change was -0.6 (-7.4 to 5.3) with R223+P and -1.3 (-3.1 to 4.8) with R223 (P = 0.66). Median rPFS and OS was 6.1 (95% confidence interval: 2.7-11.0) and 16.9 months [12.7-not reached (NR)], respectively, with R223+P and 5.7 (2.6-NR) and 16.0 (9.0-NR), respectively, with R223. Although R223+P was well tolerated with no unexpected toxicity, the combination did not improve efficacy. High-dimensional flow cytometry demonstrated minimal immune modulation with R223, whereas R223+P induced CTLA-4 expression on circulating CD4+ T cells. Clinical responders possessed lower circulating frequencies of Ki67+ T and myeloid cells at baseline and higher circulating frequencies of TIM-3+ T and myeloid cells by week 9. Although R223+P did not induce T-cell infiltration into the tumor microenvironment, exhaustion of induced peripheral T-cell immune responses may dampen the combination's clinical activity.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"704-718"},"PeriodicalIF":8.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11148544/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140326365","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A Sampling of Highlights from the Literature. 文献精华选集》。
IF 10.1 1区 医学
Cancer immunology research Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-12-6-WWR
{"title":"A Sampling of Highlights from the Literature.","authors":"","doi":"10.1158/2326-6066.CIR-12-6-WWR","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-12-6-WWR","url":null,"abstract":"","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":"12 6","pages":"651"},"PeriodicalIF":10.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141236956","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
An IRF2-Expressing Oncolytic Virus Changes the Susceptibility of Tumor Cells to Antitumor T Cells and Promotes Tumor Clearance. 表达 IRF2 的溶瘤病毒可改变肿瘤细胞对抗肿瘤 T 细胞的易感性并促进肿瘤清除
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2024-06-04 DOI: 10.1158/2326-6066.CIR-23-0573
Lulu Shao, Rashmi Srivastava, Greg M Delgoffe, Stephen H Thorne, Saumendra N Sarkar
{"title":"An IRF2-Expressing Oncolytic Virus Changes the Susceptibility of Tumor Cells to Antitumor T Cells and Promotes Tumor Clearance.","authors":"Lulu Shao, Rashmi Srivastava, Greg M Delgoffe, Stephen H Thorne, Saumendra N Sarkar","doi":"10.1158/2326-6066.CIR-23-0573","DOIUrl":"10.1158/2326-6066.CIR-23-0573","url":null,"abstract":"<p><p>IFN regulatory factor 1 (IRF1) can promote antitumor immunity. However, we have shown previously that in the tumor cell, IRF1 can promote tumor growth, and IRF1-deficient tumor cells exhibit severely restricted tumor growth in several syngeneic mouse tumor models. Here, we investigate the potential of functionally modulating IRF1 to reduce tumor progression and prolong survival. Using inducible IRF1 expression, we established that it is possible to regulate IRF1 expression to modulate tumor progression in established B16-F10 tumors. Expression of IRF2, which is a functional antagonist of IRF1, downregulated IFNγ-induced expression of inhibitory ligands, upregulated MHC-related molecules, and slowed tumor growth and extended survival. We characterized the functional domain(s) of IRF2 needed for this antitumor activity, showing that a full-length IRF2 was required for its antitumor functions. Finally, using an oncolytic vaccinia virus as a delivery platform, we showed that IRF2-expressing vaccinia virus suppressed tumor progression and prolonged survival in multiple tumor models. These results suggest the potency of targeting IRF1 and using IRF2 to modulate immunotherapy.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"779-790"},"PeriodicalIF":8.1,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11150089/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140183844","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Metabolic reprogramming of tumor-associated macrophages using glutamine antagonist JHU083 drives tumor immunity in myeloid-rich prostate and bladder cancer tumors 利用谷氨酰胺拮抗剂 JHU083 对肿瘤相关巨噬细胞进行代谢重编程,促进富含骨髓的前列腺癌和膀胱癌肿瘤的肿瘤免疫功能
IF 10.1 1区 医学
Cancer immunology research Pub Date : 2024-05-03 DOI: 10.1158/2326-6066.cir-23-1105
Monali Praharaj, Fan Shen, Alex J. Lee, Liang Zhao, Thomas R. Nirschl, Debebe Theodros, Alok K. Singh, Xiaoxu Wang, Kenneth M. Adusei, Kara A. Lombardo, Raekwon A. Williams, Laura A. Sena, Elizabeth A. Thompson, Ada Tam, Srinivasan Yegnasubramanian, Edward J. Pearce, Robert D. Leone, Jesse Alt, Rana Rais, Barbara S. Slusher, Drew M. Pardoll, Jonathan D. Powell, Jelani C. Zarif
{"title":"Metabolic reprogramming of tumor-associated macrophages using glutamine antagonist JHU083 drives tumor immunity in myeloid-rich prostate and bladder cancer tumors","authors":"Monali Praharaj, Fan Shen, Alex J. Lee, Liang Zhao, Thomas R. Nirschl, Debebe Theodros, Alok K. Singh, Xiaoxu Wang, Kenneth M. Adusei, Kara A. Lombardo, Raekwon A. Williams, Laura A. Sena, Elizabeth A. Thompson, Ada Tam, Srinivasan Yegnasubramanian, Edward J. Pearce, Robert D. Leone, Jesse Alt, Rana Rais, Barbara S. Slusher, Drew M. Pardoll, Jonathan D. Powell, Jelani C. Zarif","doi":"10.1158/2326-6066.cir-23-1105","DOIUrl":"https://doi.org/10.1158/2326-6066.cir-23-1105","url":null,"abstract":"Glutamine metabolism in tumor microenvironments critically regulates anti-tumor immunity. Using glutamine-antagonist prodrug JHU083, we report potent tumor growth inhibition in urologic tumors by JHU083-reprogrammed tumor-associated macrophages (TAMs) and tumor-infiltrating monocytes (TIMs). We show JHU083-mediated glutamine antagonism in tumor microenvironments induces TNF, pro-inflammatory, and mTORC1 signaling in intratumoral TAM clusters. JHU083-reprogrammed TAMs also exhibit increased tumor cell phagocytosis and diminished pro-angiogenic capacities. In vivo inhibition of TAM glutamine consumption resulted in increased glycolysis, a broken TCA cycle, and purine metabolism disruption. Although the anti-tumor effect of glutamine antagonism on tumor-infiltrating T cells was moderate, JHU083 promoted a stem cell-like phenotype in CD8+ T cells and decreased Treg abundance. Finally, JHU083 caused a ubiquitous shutdown in glutamine utilizing metabolic pathways in tumor cells, leading to reduced HIF-1alpha, c-MYC phosphorylation, and induction of tumor cell apoptosis, all key anti-tumor features.","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":"12 1","pages":""},"PeriodicalIF":10.1,"publicationDate":"2024-05-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140841852","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
High-Dimensional Analyses Reveal IL15 Enhances Activation of Sipuleucel-T Lymphocyte Subsets and Reverses Immunoresistance. 高维分析表明,IL-15 可增强 Sipuleucel-T 淋巴细胞亚群的活化并逆转免疫阻抗。
IF 10.1 1区 医学
Cancer immunology research Pub Date : 2024-05-02 DOI: 10.1158/2326-6066.CIR-23-0652
Muhammad A Saeed, Bo Peng, Kevin Kim, Kavita Rawat, Lindsey M Kuehm, Zoe R Siegel, Ariel Borkowski, Nabih Habib, Brian Van Tine, Nadeem Sheikh, Vu Tuyen, Daniel L J Thorek, Todd A Fehniger, Russell K Pachynski
{"title":"High-Dimensional Analyses Reveal IL15 Enhances Activation of Sipuleucel-T Lymphocyte Subsets and Reverses Immunoresistance.","authors":"Muhammad A Saeed, Bo Peng, Kevin Kim, Kavita Rawat, Lindsey M Kuehm, Zoe R Siegel, Ariel Borkowski, Nabih Habib, Brian Van Tine, Nadeem Sheikh, Vu Tuyen, Daniel L J Thorek, Todd A Fehniger, Russell K Pachynski","doi":"10.1158/2326-6066.CIR-23-0652","DOIUrl":"10.1158/2326-6066.CIR-23-0652","url":null,"abstract":"<p><p>Sipuleucel-T (sip-T) is the only FDA-approved autologous cellular immunotherapy for metastatic castration-resistant prostate cancer (mCRPC). To elucidate parameters of the response profile to this therapy, we report high-dimensional analyses of sip-T using cytometry by time of flight (CyTOF) and show a lymphoid predominance, with CD3+ T cells constituting the highest proportion (median ∼60%) of sip-T, followed by B cells, and natural killer (NK) and NKT cells. We hypothesized that treatment of sip-T with homeostatic cytokines known to activate/expand effector lymphocytes could augment efficacy against prostate tumors. Of the cytokines tested, IL15 was the most effective at enhancing activation and proliferation of effector lymphocytes, as well as augmenting tumor cytotoxicity in vitro. Co-culture of sip-T with IL15 and control or prostate-relevant antigens showed substantial activation and expansion of CD8+ T cells and NKT cells in an antigen-specific manner. Adoptive transfer of IL15-treated sip-T into NSG mice resulted in more potent prostate tumor growth inhibition compared with control sip-T. Evaluation of tumor-infiltrating lymphocytes revealed a 2- to 14-fold higher influx of sip-T and a significant increase in IFNγ producing CD8+ T cells and NKT cells within the tumor microenvironment in the IL15 group. In conclusion, we put forward evidence that IL15 treatment can enhance the functional antitumor immunity of sip-T, providing rationale for combining IL15 or IL15 agonists with sip-T to treat patients with mCRPC.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"559-574"},"PeriodicalIF":10.1,"publicationDate":"2024-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139971035","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
Tsyn-Seq: a T-cell Synapse-Based Antigen Identification Platform. Tsyn-seq:基于 T 细胞突触的抗原识别平台。
IF 8.1 1区 医学
Cancer immunology research Pub Date : 2024-05-02 DOI: 10.1158/2326-6066.CIR-23-0467
Yimei Jin, Takahiko Miyama, Alexandria Brown, Tomo Hayase, Xingzhi Song, Anand K Singh, Licai Huang, Ivonne I Flores, Lauren K McDaniel, Israel Glover, Taylor M Halsey, Rishika Prasad, Valerie Chapa, Saira Ahmed, Jianhua Zhang, Kunal Rai, Christine B Peterson, Gregory Lizee, Jennifer Karmouch, Eiko Hayase, Jeffrey J Molldrem, Chia-Chi Chang, Wen-Bin Tsai, Robert R Jenq
{"title":"Tsyn-Seq: a T-cell Synapse-Based Antigen Identification Platform.","authors":"Yimei Jin, Takahiko Miyama, Alexandria Brown, Tomo Hayase, Xingzhi Song, Anand K Singh, Licai Huang, Ivonne I Flores, Lauren K McDaniel, Israel Glover, Taylor M Halsey, Rishika Prasad, Valerie Chapa, Saira Ahmed, Jianhua Zhang, Kunal Rai, Christine B Peterson, Gregory Lizee, Jennifer Karmouch, Eiko Hayase, Jeffrey J Molldrem, Chia-Chi Chang, Wen-Bin Tsai, Robert R Jenq","doi":"10.1158/2326-6066.CIR-23-0467","DOIUrl":"10.1158/2326-6066.CIR-23-0467","url":null,"abstract":"<p><p>Tools for genome-wide rapid identification of peptide-major histocompatibility complex targets of T-cell receptors (TCR) are not yet universally available. We present a new antigen screening method, the T-synapse (Tsyn) reporter system, which includes antigen-presenting cells (APC) with a Fas-inducible NF-κB reporter and T cells with a nuclear factor of activated T cells (NFAT) reporter. To functionally screen for target antigens from a cDNA library, productively interacting T cell-APC aggregates were detected by dual-reporter activity and enriched by flow sorting followed by antigen identification quantified by deep sequencing (Tsyn-seq). When applied to a previously characterized TCR specific for the E7 antigen derived from human papillomavirus type 16 (HPV16), Tsyn-seq successfully enriched the correct cognate antigen from a cDNA library derived from an HPV16-positive cervical cancer cell line. Tsyn-seq provides a method for rapidly identifying antigens recognized by TCRs of interest from a tumor cDNA library. See related Spotlight by Makani and Joglekar, p. 515.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"530-543"},"PeriodicalIF":8.1,"publicationDate":"2024-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11065584/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139740415","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
T-cell expression of CXCL13 is associated with immunotherapy response in a sex-dependent manner in patients with lung cancer 肺癌患者 T 细胞中 CXCL13 的表达与免疫疗法反应的性别依赖性有关
IF 10.1 1区 医学
Cancer immunology research Pub Date : 2024-05-02 DOI: 10.1158/2326-6066.cir-23-0826
Michelle Brennan, David DeBruin, Chinye Nwokolo, Katey S. Hunt, Alexander Piening, Maureen J. Donlin, Stephen T. Ferris, Ryan M. Teague, Richard J. DiPaolo, Elise Alspach
{"title":"T-cell expression of CXCL13 is associated with immunotherapy response in a sex-dependent manner in patients with lung cancer","authors":"Michelle Brennan, David DeBruin, Chinye Nwokolo, Katey S. Hunt, Alexander Piening, Maureen J. Donlin, Stephen T. Ferris, Ryan M. Teague, Richard J. DiPaolo, Elise Alspach","doi":"10.1158/2326-6066.cir-23-0826","DOIUrl":"https://doi.org/10.1158/2326-6066.cir-23-0826","url":null,"abstract":"Emerging evidence in preclinical models demonstrates that antitumor immunity is not equivalent between males and females. However, more investigation in patients and across a wider range of cancer types is needed to fully understand sex as a variable in tumor immune responses. We investigated differences in T-cell responses between male and female patients with lung cancer by performing sex-based analysis of single cell transcriptomic datasets. We found that the transcript encoding CXC motif chemokine ligand 13 (CXCL13), which has recently been shown to correlate with T-cell tumor specificity, is expressed at greater levels in T cells isolated from female compared to male patients. Furthermore, increased expression of CXCL13 was associated with response to PD-1–targeting immunotherapy in female but not male patients. These findings suggest that there are sex-based differences in T-cell function required for response to anti–PD-1 therapy in lung cancer that may need to be considered during patient treatment decisions.","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":"84 1","pages":""},"PeriodicalIF":10.1,"publicationDate":"2024-05-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140832690","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