首页 > 最新文献

Cancer immunology research最新文献

英文 中文
PKCδ germline variants and genetic deletion in mice augment antitumor immunity through regulation of myeloid cells.
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-14 DOI: 10.1158/2326-6066.CIR-23-0999
Kyle R Cron, Ayelet Sivan, Keston Aquino-Michaels, Andrea Ziblat, Emily F Higgs, Randy F Sweis, Ruxandra Tonea, Seoho Lee, Thomas F Gajewski

Based on the notion that hypomorphic germline genetic variants are linked to autoimmune diseases, we reasoned that novel targets for cancer immunotherapy might be identified through germline variants associated with greater T-cell infiltration into tumors. Here, we report that while investigating germline polymorphisms associated with a tumor immune gene signature, we identified PKCδ as a candidate. Genetic deletion of PKCδ in mice resulted in improved endogenous antitumor immunity and increased efficacy of anti-PD-L1. Single-cell RNA sequencing revealed myeloid cell expression of Prkcd, and PKCδ deletion caused a shift in macrophage gene expression from an M2-like to an M1-like phenotype. Conditional deletion of PKCδ in myeloid cells recapitulated improved tumor control that was augmented further with anti-PD-L1. Analysis of clinical samples confirmed an association between PRKCD variants and M1/M2 phenotype, as well as between a PKCδ KO-like gene signature and clinical benefit from anti-PD-1. Our results identify PKCδ as a candidate therapeutic target that modulates myeloid cell states.

{"title":"PKCδ germline variants and genetic deletion in mice augment antitumor immunity through regulation of myeloid cells.","authors":"Kyle R Cron, Ayelet Sivan, Keston Aquino-Michaels, Andrea Ziblat, Emily F Higgs, Randy F Sweis, Ruxandra Tonea, Seoho Lee, Thomas F Gajewski","doi":"10.1158/2326-6066.CIR-23-0999","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-23-0999","url":null,"abstract":"<p><p>Based on the notion that hypomorphic germline genetic variants are linked to autoimmune diseases, we reasoned that novel targets for cancer immunotherapy might be identified through germline variants associated with greater T-cell infiltration into tumors. Here, we report that while investigating germline polymorphisms associated with a tumor immune gene signature, we identified PKCδ as a candidate. Genetic deletion of PKCδ in mice resulted in improved endogenous antitumor immunity and increased efficacy of anti-PD-L1. Single-cell RNA sequencing revealed myeloid cell expression of Prkcd, and PKCδ deletion caused a shift in macrophage gene expression from an M2-like to an M1-like phenotype. Conditional deletion of PKCδ in myeloid cells recapitulated improved tumor control that was augmented further with anti-PD-L1. Analysis of clinical samples confirmed an association between PRKCD variants and M1/M2 phenotype, as well as between a PKCδ KO-like gene signature and clinical benefit from anti-PD-1. Our results identify PKCδ as a candidate therapeutic target that modulates myeloid cell states.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.1,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142977624","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
In situ detection of individual classical MHC-I gene products in cancer.
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-13 DOI: 10.1158/2326-6066.CIR-24-1003
Paula I Gonzalez-Ericsson, Susan R Opalenik, Violeta Sanchez, Amy Palubinsky, Ann Hanna, Xiaopeng Sun, Andres A Ocampo, Guadalupe Garcia, Leonel Maldonado, Zaida Morante, Tatiana Vidaurre, Guillermo Valencia, Henry L Gomez, Melinda E Sanders, Laura C Kennedy, Elizabeth Phillips, Justin M Balko

Tumor-specific HLA class I expression is required for cytotoxic T-cell elimination of cancer cells expressing tumor-associated or neo-antigens. Cancers downregulate antigen presentation to avoid adaptive immunity. The highly polymorphic nature of the genes encoding these proteins, coupled with quaternary-structure changes after formalin fixation, complicate detection by immunohistochemistry. In this study we determined recognition of 16 specific HLA-A, B and C alleles by 15 antibodies commercially available for immunohistochemical use, identifying and validating pan and specific HLA-A, B, and C antibodies, providing a validated method that can be applied to investigate HLA-A, B and C molecule-specific loss in cancer. We applied this approach to a series of breast cancers as a proof of utility, identifying differential HLA-A, B and C loss, with a higher incidence of HLA-A and B loss in hormone-driven breast cancers, HLA-B loss in HER2+ cancers, and an equal loss of all three molecules in triple-negative disease. Additionally, we found that at the protein level, HLA-A and B loss were early events prevalent in premalignant lesions, while HLA-C loss was less common throughout tumor evolution. Effective response to immunotherapies such as checkpoint inhibitors and MHC-I-targeted cancer vaccines, which hinge on the carriage of specific allele groups, require MHC-I expression on tumor cells. These findings have implications for the success of checkpoint inhibitors and vaccine strategies.

{"title":"In situ detection of individual classical MHC-I gene products in cancer.","authors":"Paula I Gonzalez-Ericsson, Susan R Opalenik, Violeta Sanchez, Amy Palubinsky, Ann Hanna, Xiaopeng Sun, Andres A Ocampo, Guadalupe Garcia, Leonel Maldonado, Zaida Morante, Tatiana Vidaurre, Guillermo Valencia, Henry L Gomez, Melinda E Sanders, Laura C Kennedy, Elizabeth Phillips, Justin M Balko","doi":"10.1158/2326-6066.CIR-24-1003","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-24-1003","url":null,"abstract":"<p><p>Tumor-specific HLA class I expression is required for cytotoxic T-cell elimination of cancer cells expressing tumor-associated or neo-antigens. Cancers downregulate antigen presentation to avoid adaptive immunity. The highly polymorphic nature of the genes encoding these proteins, coupled with quaternary-structure changes after formalin fixation, complicate detection by immunohistochemistry. In this study we determined recognition of 16 specific HLA-A, B and C alleles by 15 antibodies commercially available for immunohistochemical use, identifying and validating pan and specific HLA-A, B, and C antibodies, providing a validated method that can be applied to investigate HLA-A, B and C molecule-specific loss in cancer. We applied this approach to a series of breast cancers as a proof of utility, identifying differential HLA-A, B and C loss, with a higher incidence of HLA-A and B loss in hormone-driven breast cancers, HLA-B loss in HER2+ cancers, and an equal loss of all three molecules in triple-negative disease. Additionally, we found that at the protein level, HLA-A and B loss were early events prevalent in premalignant lesions, while HLA-C loss was less common throughout tumor evolution. Effective response to immunotherapies such as checkpoint inhibitors and MHC-I-targeted cancer vaccines, which hinge on the carriage of specific allele groups, require MHC-I expression on tumor cells. These findings have implications for the success of checkpoint inhibitors and vaccine strategies.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.1,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142969016","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
Preclinical Development of T Cells Engineered to Express a T-Cell Antigen Coupler Targeting Claudin 18.2-Positive Solid Tumors. 针对 Claudin 18.2 阳性实体瘤设计表达 T 细胞抗原偶联剂 (TAC) 的 T 细胞的临床前开发。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-09 DOI: 10.1158/2326-6066.CIR-24-0138
Stacey X Xu, Ling Wang, Philbert Ip, Ritu R Randhawa, Tania Benatar, Suzanna L Prosser, Prabha Lal, Alima Naim Khan, Thanyashanthi Nitya-Nootan, Gargi Thakor, Heather MacGregor, Danielle L Hayes, Andrea Vucicevic, Princy Mathew, Sadhak Sengupta, Christopher W Helsen, Andreas G Bader

The T-cell antigen coupler (TAC) is a chimeric receptor that facilitates tumor antigen-specific activation of T cells by co-opting the endogenous T-cell receptor complex in the absence of tonic signaling. Previous data demonstrate that the TAC affords T cells with the ability to induce durable and safe antitumor responses in preclinical models of hematologic and solid tumors. In this study, we describe the preclinical pharmacology and safety of an autologous Claudin 18.2 (CLDN18.2)-directed TAC T-cell therapy, TAC01-CLDN18.2, in preparation for a phase I/II clinical study in subjects with CLDN18.2-positive solid tumors. Following a screen of putative TAC constructs, the specificity, activity, and cytotoxicity of TAC T cells expressing the final CLDN18.2-TAC receptor were evaluated in vitro and in vivo using gastric, gastroesophageal, and pancreatic tumor models as well as human cells derived from normal tissues. CLDN18.2-specific activity and cytotoxicity of CLDN18.2-TAC T cells were observed in coculture with various 2D tumor cultures naturally expressing CLDN18.2 as well as tumor spheroids. These effects occurred in models with low antigen levels and were positively associated with increasing CLDN18.2 expression. CLDN18.2-TAC T cells effectively eradicated established tumor xenografts in mice in the absence of observed off-target or on-target/off-tumor effects, elicited durable efficacy in recursive killing and tumor rechallenge experiments, and remained unreactive in coculture with human cells representing vital organs. Thus, the data demonstrate that CLDN18.2-TAC T cells can induce a specific and long-lasting antitumor response in various CLDN18.2-positive solid tumor models without notable TAC-dependent toxicities, supporting the clinical development of TAC01-CLDN18.2.

T 细胞抗原偶联体(TAC)是一种嵌合受体,它能在没有补体信号的情况下通过与内源性 T 细胞受体复合物共用来促进肿瘤抗原特异性激活 T 细胞。以前的数据表明,TAC 使 T 细胞有能力在血液肿瘤和实体肿瘤的临床前模型中诱导持久、安全的抗肿瘤反应。在这里,我们描述了一种自体Claudin 18.2(CLDN18.2)导向的TAC T细胞疗法TAC01-CLDN18.2的临床前药理学和安全性,为在CLDN18.2阳性实体瘤受试者中进行I/II期临床研究做准备。在对可能的 TAC 构建物进行筛选后,利用胃、胃食管和胰腺肿瘤模型以及来自正常组织的人体细胞对表达最终 CLDN18.2-TAC 受体的 TAC T 细胞的特异性、活性和细胞毒性进行了体外和体内评估。在与各种天然表达 CLDN18.2 的二维肿瘤培养物以及肿瘤球状体共培养时,观察到了 CLDN18.2-TAC T 细胞的 CLDN18.2 特异性活性和细胞毒性。这些效应发生在抗原水平较低的模型中,并与 CLDN18.2 表达的增加呈正相关。CLDN18.2-TAC T 细胞在没有观察到脱靶或靶上/脱瘤效应的情况下有效清除了小鼠体内已建立的肿瘤异种移植物,在复发杀伤和肿瘤再挑战实验中产生了持久的疗效,并且在与代表重要器官的人体细胞共培养时仍无反应。因此,这些数据表明,CLDN18.2-TAC T 细胞能在各种 CLDN18.2 阳性实体瘤模型中诱导特异性和持久的抗肿瘤反应,且无明显的 TAC 依赖性毒性,支持 TAC01-CLDN18.2 的临床开发。
{"title":"Preclinical Development of T Cells Engineered to Express a T-Cell Antigen Coupler Targeting Claudin 18.2-Positive Solid Tumors.","authors":"Stacey X Xu, Ling Wang, Philbert Ip, Ritu R Randhawa, Tania Benatar, Suzanna L Prosser, Prabha Lal, Alima Naim Khan, Thanyashanthi Nitya-Nootan, Gargi Thakor, Heather MacGregor, Danielle L Hayes, Andrea Vucicevic, Princy Mathew, Sadhak Sengupta, Christopher W Helsen, Andreas G Bader","doi":"10.1158/2326-6066.CIR-24-0138","DOIUrl":"10.1158/2326-6066.CIR-24-0138","url":null,"abstract":"<p><p>The T-cell antigen coupler (TAC) is a chimeric receptor that facilitates tumor antigen-specific activation of T cells by co-opting the endogenous T-cell receptor complex in the absence of tonic signaling. Previous data demonstrate that the TAC affords T cells with the ability to induce durable and safe antitumor responses in preclinical models of hematologic and solid tumors. In this study, we describe the preclinical pharmacology and safety of an autologous Claudin 18.2 (CLDN18.2)-directed TAC T-cell therapy, TAC01-CLDN18.2, in preparation for a phase I/II clinical study in subjects with CLDN18.2-positive solid tumors. Following a screen of putative TAC constructs, the specificity, activity, and cytotoxicity of TAC T cells expressing the final CLDN18.2-TAC receptor were evaluated in vitro and in vivo using gastric, gastroesophageal, and pancreatic tumor models as well as human cells derived from normal tissues. CLDN18.2-specific activity and cytotoxicity of CLDN18.2-TAC T cells were observed in coculture with various 2D tumor cultures naturally expressing CLDN18.2 as well as tumor spheroids. These effects occurred in models with low antigen levels and were positively associated with increasing CLDN18.2 expression. CLDN18.2-TAC T cells effectively eradicated established tumor xenografts in mice in the absence of observed off-target or on-target/off-tumor effects, elicited durable efficacy in recursive killing and tumor rechallenge experiments, and remained unreactive in coculture with human cells representing vital organs. Thus, the data demonstrate that CLDN18.2-TAC T cells can induce a specific and long-lasting antitumor response in various CLDN18.2-positive solid tumor models without notable TAC-dependent toxicities, supporting the clinical development of TAC01-CLDN18.2.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"35-46"},"PeriodicalIF":8.1,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11712040/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142458700","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
TBK1 Targeting Is Identified as a Therapeutic Strategy to Enhance CAR T-Cell Efficacy Using Patient-Derived Organotypic Tumor Spheroids.
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-09 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

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.

{"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":"https://doi.org/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":"OF1-OF19"},"PeriodicalIF":8.1,"publicationDate":"2025-01-09","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
Gefitinib Reverses PD-L1-Mediated Immunosuppression Induced by Long-term Glutamine Blockade in Bladder Cancer. 吉非替尼能逆转PD-L1介导的膀胱癌谷氨酰胺长期阻断诱导的免疫抑制
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-09 DOI: 10.1158/2326-6066.CIR-24-0039
Guofeng Ma, Huiqing Jia, Zhiqiang Li, Xiangyan Zhang, Liping Wang, Zhilei Zhang, Yujing Xiao, Zhijuan Liang, Dan Li, Yuanbin Chen, Xintao Tian, Yonghua Wang, Ye Liang, Haitao Niu

Glutamine is a major energy source for tumor cells, and blocking glutamine metabolism is being investigated as a promising strategy for cancer therapy. However, the antitumor effect of glutamine blockade in bladder cancer remains unclear, necessitating further investigation. In this study, we demonstrated that glutamine metabolism was involved in the malignant progression of bladder cancer. Treatment with the glutamine antagonist 6-diazo-5-oxo-L-norleucine (DON) inhibited the growth of bladder cancer cells in vitro in several ways. In addition, we observed inhibition of tumor growth in bladder cancer-bearing mice by using JHU083, a prodrug that was designed to prevent DON-induced toxicity. However, the antitumor immune effect of T cells changed from activation to inhibition as the administrated time extended. We found that both in vitro treatment with DON and in vivo prolonged administration of JHU083 led to the upregulation of PD-L1 in bladder cancer cells. Mechanistically, glutamine blockade upregulated PD-L1 expression in bladder cancer cells by accumulating reactive oxygen species, subsequently activating the EGFR/ERK/C-Jun signaling pathway. Combination treatment of JHU083 and gefitinib reversed the upregulation of PD-L1 in bladder cancer cells induced by prolonged glutamine blockade, resulting in the alleviation of T-cell immunosuppression and a significant improvement in therapeutic outcome. These preclinical findings show promise for glutamine metabolism targeting as a viable therapeutic strategy for bladder cancer, with the potential for further enhancement through combined treatment with gefitinib.

谷氨酰胺是肿瘤细胞的主要能量来源,阻断谷氨酰胺代谢作为一种很有前景的癌症治疗策略正在接受研究。然而,谷氨酰胺在膀胱癌中的抗肿瘤作用仍不明确,需要进一步研究。在这里,我们证实谷氨酰胺代谢参与了膀胱癌的恶性进展。谷氨酰胺拮抗剂 6-重氮-5-氧代-L-正亮氨酸(DON)通过多种方式抑制体外膀胱癌细胞的生长。此外,我们还观察到使用 JHU083(一种用于防止 DON 引起的毒性的原药)对膀胱癌小鼠肿瘤生长的抑制作用。然而,随着给药时间的延长,T 细胞的抗肿瘤免疫效应从激活变为抑制。我们发现,体外使用 DON 和体内长期服用 JHU083 都会导致膀胱癌细胞中 PD-L1 的上调。从机制上讲,谷氨酰胺阻断通过积累 ROS 上调膀胱癌细胞中 PD-L1 的表达,继而激活表皮生长因子受体/ERK/C-Jun 信号通路。JHU083和吉非替尼的联合治疗逆转了谷氨酰胺长期阻断诱导的膀胱癌细胞中PD-L1的上调,从而缓解了T细胞免疫抑制,显著改善了治疗效果。这些临床前研究结果表明,谷氨酰胺代谢靶向有望成为一种可行的膀胱癌治疗策略,并有可能通过与吉非替尼联合治疗进一步提高疗效。
{"title":"Gefitinib Reverses PD-L1-Mediated Immunosuppression Induced by Long-term Glutamine Blockade in Bladder Cancer.","authors":"Guofeng Ma, Huiqing Jia, Zhiqiang Li, Xiangyan Zhang, Liping Wang, Zhilei Zhang, Yujing Xiao, Zhijuan Liang, Dan Li, Yuanbin Chen, Xintao Tian, Yonghua Wang, Ye Liang, Haitao Niu","doi":"10.1158/2326-6066.CIR-24-0039","DOIUrl":"10.1158/2326-6066.CIR-24-0039","url":null,"abstract":"<p><p>Glutamine is a major energy source for tumor cells, and blocking glutamine metabolism is being investigated as a promising strategy for cancer therapy. However, the antitumor effect of glutamine blockade in bladder cancer remains unclear, necessitating further investigation. In this study, we demonstrated that glutamine metabolism was involved in the malignant progression of bladder cancer. Treatment with the glutamine antagonist 6-diazo-5-oxo-L-norleucine (DON) inhibited the growth of bladder cancer cells in vitro in several ways. In addition, we observed inhibition of tumor growth in bladder cancer-bearing mice by using JHU083, a prodrug that was designed to prevent DON-induced toxicity. However, the antitumor immune effect of T cells changed from activation to inhibition as the administrated time extended. We found that both in vitro treatment with DON and in vivo prolonged administration of JHU083 led to the upregulation of PD-L1 in bladder cancer cells. Mechanistically, glutamine blockade upregulated PD-L1 expression in bladder cancer cells by accumulating reactive oxygen species, subsequently activating the EGFR/ERK/C-Jun signaling pathway. Combination treatment of JHU083 and gefitinib reversed the upregulation of PD-L1 in bladder cancer cells induced by prolonged glutamine blockade, resulting in the alleviation of T-cell immunosuppression and a significant improvement in therapeutic outcome. These preclinical findings show promise for glutamine metabolism targeting as a viable therapeutic strategy for bladder cancer, with the potential for further enhancement through combined treatment with gefitinib.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"66-83"},"PeriodicalIF":8.1,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142521079","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
Alternate MHC I Antigen Presentation Pathways Allow CD8+ T-cell Recognition and Killing of Cancer Cells in the Absence of β2M or TAP. 在没有 ß2M 或 TAP 的情况下,替代的 MHC I 抗原递呈途径可使 CD8+ T 细胞识别并杀死癌细胞。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-09 DOI: 10.1158/2326-6066.CIR-24-0320
Freidrich M Cruz, Laura A A Orellano, Amanda Chan, Kenneth L Rock

MHC I antigen presentation allows CD8+ T cells to detect and eliminate cancerous or virally infected cells. The MHC I pathway is not essential for cell growth and viability, so cancers and viruses can evade control by CD8+ T cells by inactivating antigen presentation. In cancers, two common ways for this evasion are the loss of either the MHC I light chain [β2 microglobulin (β2M)] or the transporter-associated with antigen processing (TAP). β2M-null cells are generally thought to lack the MHC I pathway because the MHC I heavy chain by itself lacks the proper conformation for peptide display. TAP-null cells are thought to have severely defective MHC I antigen presentation because they are incapable of supplying peptides from the cytosol to MHC I molecules in the endoplasmic reticulum (ER). However, we have found that highly reactive memory CD8+ T cells could still recognize cells that completely lacked β2M or TAP. This was at least in part because in TAP-null cells, the Sec62 component of the Sec61 translocon supported the transfer of cytosolic peptides into the ER. In β2M-negative cells, free MHC I heavy chains were able to bind peptides and assume a conformation that was sufficiently recognized by CD8+ T cells. This process required ER chaperones and the peptide-loading complex. We found that these mechanisms supported antigen presentation at a level that was sufficient for memory CD8+ T cells to kill melanoma cells both in vitro and in tumor-bearing mice. The implications for tumor immunotherapy are discussed.

主要组织蛋白酶复合物 I 类(MHC I)抗原呈递允许 CD8+ T 细胞检测并消灭癌细胞或受病毒感染的细胞。MHC I 途径并非细胞生长和存活所必需,因此癌症和病毒可以通过使抗原递呈失活来逃避 CD8+ T 细胞的控制。在癌症中,有两种常见的逃避方式,一种是失去 MHC I 轻链(ß2M),另一种是失去细胞质到内质网(ER)的多肽转运体(TAP)。一般认为 ß2M 缺失的细胞缺乏 MHC I 途径,因为 MHC I 重链本身缺乏肽显示的适当构象。TAP无效细胞被认为具有严重的MHC I抗原呈递缺陷,因为它们无法从细胞质向ER中的MHC I分子提供肽。然而,我们发现,高活性记忆CD8+ T细胞仍能识别完全缺乏ß2M或TAP的细胞。这至少部分是因为在TAP缺失的细胞中,Sec61转座子的Sec62成分支持将细胞膜肽转移到ER中。在ß2M阴性细胞中,游离的MHC I重链能够与肽结合,并形成足以被CD8+ T细胞识别的构象。这一过程需要ER伴侣和多肽装载复合体。我们发现,这些机制支持的抗原呈递水平足以让记忆 CD8+ T 细胞在体外和肿瘤小鼠体内杀死黑色素瘤细胞。本文讨论了这一机制对肿瘤免疫疗法的影响。
{"title":"Alternate MHC I Antigen Presentation Pathways Allow CD8+ T-cell Recognition and Killing of Cancer Cells in the Absence of β2M or TAP.","authors":"Freidrich M Cruz, Laura A A Orellano, Amanda Chan, Kenneth L Rock","doi":"10.1158/2326-6066.CIR-24-0320","DOIUrl":"10.1158/2326-6066.CIR-24-0320","url":null,"abstract":"<p><p>MHC I antigen presentation allows CD8+ T cells to detect and eliminate cancerous or virally infected cells. The MHC I pathway is not essential for cell growth and viability, so cancers and viruses can evade control by CD8+ T cells by inactivating antigen presentation. In cancers, two common ways for this evasion are the loss of either the MHC I light chain [β2 microglobulin (β2M)] or the transporter-associated with antigen processing (TAP). β2M-null cells are generally thought to lack the MHC I pathway because the MHC I heavy chain by itself lacks the proper conformation for peptide display. TAP-null cells are thought to have severely defective MHC I antigen presentation because they are incapable of supplying peptides from the cytosol to MHC I molecules in the endoplasmic reticulum (ER). However, we have found that highly reactive memory CD8+ T cells could still recognize cells that completely lacked β2M or TAP. This was at least in part because in TAP-null cells, the Sec62 component of the Sec61 translocon supported the transfer of cytosolic peptides into the ER. In β2M-negative cells, free MHC I heavy chains were able to bind peptides and assume a conformation that was sufficiently recognized by CD8+ T cells. This process required ER chaperones and the peptide-loading complex. We found that these mechanisms supported antigen presentation at a level that was sufficient for memory CD8+ T cells to kill melanoma cells both in vitro and in tumor-bearing mice. The implications for tumor immunotherapy are discussed.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"98-108"},"PeriodicalIF":8.1,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11717633/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142562642","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
Level of Expression of MHCI-Presented Neoepitopes Influences Tumor Rejection by Neoantigen-Specific CD8+ T Cells. MHCI 呈递的新表位表达水平会影响新抗原特异性 CD8+ T 细胞对肿瘤的排斥反应。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-09 DOI: 10.1158/2326-6066.CIR-23-0639
Li Deng, Scott R Walsh, Andrew Nguyen, Jordon M Inkol, Michael J Westerveld, Lan Chen, Nader El-Sayes, Karen L Mossman, Samuel T Workenhe, Yonghong Wan

Neoantigen-targeted therapy holds an array of benefits for cancer immunotherapy, but the identification of peptide targets with tumor rejection capacity remains a limitation. To better define the criteria dictating tumor rejection potential, we examined the capacity of high-magnitude T-cell responses induced toward several distinct neoantigen targets to regress MC38 tumors. Despite their demonstrated immunogenicity, vaccine-induced T-cell responses were unable to regress established MC38 tumors or prevent tumor engraftment in a prophylactic setting. Although unable to kill tumor cells, T cells showed robust killing capacity toward neoantigen peptide-loaded cells. Tumor-cell killing was rescued by saturation of target peptide-loaded MHCs on the cell surface. Overall, this study demonstrates a pivotal role for target protein expression levels in modulating the tumor rejection capacity of neoantigens. Thus, inclusion of this metric, in addition to immunogenicity analysis, may benefit antigen prediction techniques to ensure the full antitumor effect of cancer vaccines.

新抗原靶向疗法为癌症免疫疗法带来了一系列益处,但具有肿瘤排斥能力的肽靶点的鉴定仍是一个局限。为了更好地定义决定肿瘤排斥潜力的标准,我们研究了针对几种不同的新抗原靶点诱导的高水平T细胞反应消退MC38肿瘤的能力。令人惊讶的是,尽管疫苗诱导的T细胞应答具有明显的免疫原性,但在预防性治疗中却无法消退已建立的MC38肿瘤或阻止肿瘤的移植。不过,T细胞具有功能性,对负载新抗原肽的细胞具有强大的杀伤能力。此外,肿瘤细胞杀伤力的恢复与细胞表面靶肽载体 MHC 的表达水平或饱和度成正比。总之,这项研究证明了靶蛋白表达水平在调节新抗原的肿瘤排斥能力中的关键作用。因此,除了免疫原性分析外,纳入这一指标可能会有利于抗原预测技术,以确保癌症疫苗的全面抗肿瘤效果。
{"title":"Level of Expression of MHCI-Presented Neoepitopes Influences Tumor Rejection by Neoantigen-Specific CD8+ T Cells.","authors":"Li Deng, Scott R Walsh, Andrew Nguyen, Jordon M Inkol, Michael J Westerveld, Lan Chen, Nader El-Sayes, Karen L Mossman, Samuel T Workenhe, Yonghong Wan","doi":"10.1158/2326-6066.CIR-23-0639","DOIUrl":"10.1158/2326-6066.CIR-23-0639","url":null,"abstract":"<p><p>Neoantigen-targeted therapy holds an array of benefits for cancer immunotherapy, but the identification of peptide targets with tumor rejection capacity remains a limitation. To better define the criteria dictating tumor rejection potential, we examined the capacity of high-magnitude T-cell responses induced toward several distinct neoantigen targets to regress MC38 tumors. Despite their demonstrated immunogenicity, vaccine-induced T-cell responses were unable to regress established MC38 tumors or prevent tumor engraftment in a prophylactic setting. Although unable to kill tumor cells, T cells showed robust killing capacity toward neoantigen peptide-loaded cells. Tumor-cell killing was rescued by saturation of target peptide-loaded MHCs on the cell surface. Overall, this study demonstrates a pivotal role for target protein expression levels in modulating the tumor rejection capacity of neoantigens. Thus, inclusion of this metric, in addition to immunogenicity analysis, may benefit antigen prediction techniques to ensure the full antitumor effect of cancer vaccines.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"84-97"},"PeriodicalIF":8.1,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142388319","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
The bidirectional interplay between T cell-based immunotherapies and the tumor microenvironment.
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-09 DOI: 10.1158/2326-6066.CIR-24-0857
Alfredo Pherez-Farah, Gioia Boncompagni, Aleksey Chudnovskiy, Giulia Pasqual

T cell-based therapies, including Tumor Infiltrating Lymphocyte Therapy (TIL), T cell receptor engineered T cells (TCR T), and Chimeric Antigen Receptor T cells (CAR T), are powerful therapeutic approaches for cancer treatment. While these therapies are primarily known for their direct cytotoxic effects on cancer cells, accumulating evidence indicates that they also influence the tumor microenvironment (TME), by altering the cytokine milieu and recruiting additional effector populations to help orchestrate the antitumor immune response. Conversely, the TME itself can modulate the behaviour of these therapies within the host by either supporting or inhibiting their activity. In this review we provide an overview of clinical and preclinical data on the bidirectional influences between T cell therapies and the TME. Unravelling the interactions between T cell-based therapies and the TME is critical for a better understanding of their mechanisms of action, resistance, and toxicity, with the goal of optimizing efficacy and safety.

{"title":"The bidirectional interplay between T cell-based immunotherapies and the tumor microenvironment.","authors":"Alfredo Pherez-Farah, Gioia Boncompagni, Aleksey Chudnovskiy, Giulia Pasqual","doi":"10.1158/2326-6066.CIR-24-0857","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-24-0857","url":null,"abstract":"<p><p>T cell-based therapies, including Tumor Infiltrating Lymphocyte Therapy (TIL), T cell receptor engineered T cells (TCR T), and Chimeric Antigen Receptor T cells (CAR T), are powerful therapeutic approaches for cancer treatment. While these therapies are primarily known for their direct cytotoxic effects on cancer cells, accumulating evidence indicates that they also influence the tumor microenvironment (TME), by altering the cytokine milieu and recruiting additional effector populations to help orchestrate the antitumor immune response. Conversely, the TME itself can modulate the behaviour of these therapies within the host by either supporting or inhibiting their activity. In this review we provide an overview of clinical and preclinical data on the bidirectional influences between T cell therapies and the TME. Unravelling the interactions between T cell-based therapies and the TME is critical for a better understanding of their mechanisms of action, resistance, and toxicity, with the goal of optimizing efficacy and safety.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.1,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142945094","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
Deleting Trim33 in Myeloid Cells Improves the Efficiency of Radiotherapy through an IFNβ-Dependent Antitumor Immune Response. 通过干扰素 beta 依赖性抗肿瘤免疫反应,删除骨髓细胞中的 Trim33 可提高放疗效率。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-09 DOI: 10.1158/2326-6066.CIR-24-0026
Anaïs Assouvie, Marine Gerbé-de-Thoré, Claire Torres, Véronique Ménard, Alexia Alfaro, Eric Deutsch, Michele Mondini, Germain Rousselet

Radiotherapy (RT) triggers an immune response that contributes to antitumor effects. Induction of IFNβ is a key event in this immunogenicity of RT. We have previously shown that TRIM33, a chromatin reader, restrains IFNβ expression in Toll-like receptor-activated myeloid cells. In this study, we explored whether deleting Trim33 in myeloid cells might improve the radio-induced immune response and subsequent efficiency of RT. We first established that Trim33-/- bone marrow-derived macrophages showed increased expression of IFNβ in response to direct irradiation, or to treatment with irradiated cancer cells, further supporting our hypothesis. We then tested the efficiency of a single-dose RT in three subcutaneous tumor models and one orthotopic tumor model. In all models, myeloid deletion of Trim33 led to a significantly improved response after RT, leading to a complete and durable response in most of the treated mice bearing orthotopic oral tumors. This effect required the involvement of the type I IFN pathway and the presence of CD8+ T lymphocytes but not NK cells. In addition, cured mice were capable of rejecting a secondary tumor challenge, demonstrating an in situ vaccination effect. We conclude that deleting Trim33 in myeloid cells improves RT efficiency, through a mechanism involving the type I IFN pathway and the immune response. Our work suggests that myeloid Trim33 is a host factor affecting the tumor response to RT, thus representing a new potential therapeutic target for modifying RT responses.

放疗(RT)会引发免疫反应,从而产生抗肿瘤效应。诱导干扰素 beta(IFN-β)是 RT 免疫原性的一个关键事件。我们之前已经证明,染色质阅读器 TRIM33 可抑制 IFN-β 在 Toll 样受体激活的骨髓细胞中的表达。在此,我们探讨了在髓系细胞中删除 Trim33 是否会改善放射诱导的免疫反应以及随后 RT 的效率。我们首先确定了 Trim33-/- 骨髓源性巨噬细胞在直接照射或照射癌细胞处理后 IFN-β 的表达增加,进一步支持了我们的假设。然后,我们在三个皮下肿瘤模型和一个正位肿瘤模型中测试了单剂量 RT 的效率。在所有情况下,Trim33的髓样体缺失都能显著改善RT后的反应,从而使大多数接受治疗的口腔正位肿瘤小鼠获得完全和持久的反应。这种效应需要 IFN-I 途径和 CD8+ T 淋巴细胞的存在,但不需要 NK 细胞。此外,治愈的小鼠能够拒绝二次肿瘤挑战,显示了原位疫苗接种效应。我们的结论是,通过涉及 IFN-I 通路和免疫反应的机制,删除髓系细胞中的 Trim33 可提高 RT 的效率。我们的研究表明,髓系细胞 Trim33 是影响肿瘤对 RT 反应的宿主因子,因此是改变 RT 反应的一个新的潜在治疗靶点。
{"title":"Deleting Trim33 in Myeloid Cells Improves the Efficiency of Radiotherapy through an IFNβ-Dependent Antitumor Immune Response.","authors":"Anaïs Assouvie, Marine Gerbé-de-Thoré, Claire Torres, Véronique Ménard, Alexia Alfaro, Eric Deutsch, Michele Mondini, Germain Rousselet","doi":"10.1158/2326-6066.CIR-24-0026","DOIUrl":"10.1158/2326-6066.CIR-24-0026","url":null,"abstract":"<p><p>Radiotherapy (RT) triggers an immune response that contributes to antitumor effects. Induction of IFNβ is a key event in this immunogenicity of RT. We have previously shown that TRIM33, a chromatin reader, restrains IFNβ expression in Toll-like receptor-activated myeloid cells. In this study, we explored whether deleting Trim33 in myeloid cells might improve the radio-induced immune response and subsequent efficiency of RT. We first established that Trim33-/- bone marrow-derived macrophages showed increased expression of IFNβ in response to direct irradiation, or to treatment with irradiated cancer cells, further supporting our hypothesis. We then tested the efficiency of a single-dose RT in three subcutaneous tumor models and one orthotopic tumor model. In all models, myeloid deletion of Trim33 led to a significantly improved response after RT, leading to a complete and durable response in most of the treated mice bearing orthotopic oral tumors. This effect required the involvement of the type I IFN pathway and the presence of CD8+ T lymphocytes but not NK cells. In addition, cured mice were capable of rejecting a secondary tumor challenge, demonstrating an in situ vaccination effect. We conclude that deleting Trim33 in myeloid cells improves RT efficiency, through a mechanism involving the type I IFN pathway and the immune response. Our work suggests that myeloid Trim33 is a host factor affecting the tumor response to RT, thus representing a new potential therapeutic target for modifying RT responses.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":"109-121"},"PeriodicalIF":8.1,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142342193","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
Host tissue factors predict immune surveillance and therapeutic outcomes in gastric cancer.
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2025-01-09 DOI: 10.1158/2326-6066.CIR-23-0563
Miseker Abate, Emily Stroobant, Teng Fei, Ya-Hui Lin, Shoji Shimada, Harrison Drebin, Eunise Chen, Laura H Tang, Sohrab P Shah, Jedd D Wolchok, Yelena Y Janjigian, Vivian E Strong, Santosha A Vardhana

The immune composition of solid tumors is typically inferred from biomarkers, such as histologic and molecular classifications, somatic mutational burden, and PD-L1 expression. However, the extent to which these biomarkers predict the immune landscape in gastric adenocarcinoma-an aggressive cancer often linked to chronic inflammation-remains poorly understood. We leveraged high-dimensional spectral cytometry to generate a comprehensive single-cell immune landscape of tumors, normal tissue, and lymph nodes from patients in the Western Hemisphere with gastric adenocarcinoma. The immune composition of gastric tumors could not be predicted by traditional metrics such as tumor histology, molecular classification, mutational burden, or PD-L1 expression via IHC. Instead, our findings revealed that innate immune surveillance within tumors could be anticipated by the immune profile of the normal gastric mucosa. Additionally, distinct T-cell states in the lymph nodes were linked to the accumulation of activated and memory-like CD8+ tumor-infiltrating lymphocytes (TILs). Unbiased re-classification of patients based on tumor-specific immune infiltrate generated four distinct subtypes with varying immune compositions. Tumors with a T-cell-dominant immune subtype, which spanned TCGA molecular subtypes, were exclusively associated with superior responses to immunotherapy. Parallel analysis of metastatic gastric cancer patients treated with immune checkpoint blockade showed that patients who responded to immunotherapy had a pre-treatment tumor composition that corresponded to a T-cell-dominant immune subtype from our analysis. Taken together, this work identifies key host-specific factors associated with intratumoral immune composition in gastric cancer and offers an immunological classification system that can effectively identify patients likely to benefit from immune-based therapies.

{"title":"Host tissue factors predict immune surveillance and therapeutic outcomes in gastric cancer.","authors":"Miseker Abate, Emily Stroobant, Teng Fei, Ya-Hui Lin, Shoji Shimada, Harrison Drebin, Eunise Chen, Laura H Tang, Sohrab P Shah, Jedd D Wolchok, Yelena Y Janjigian, Vivian E Strong, Santosha A Vardhana","doi":"10.1158/2326-6066.CIR-23-0563","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-23-0563","url":null,"abstract":"<p><p>The immune composition of solid tumors is typically inferred from biomarkers, such as histologic and molecular classifications, somatic mutational burden, and PD-L1 expression. However, the extent to which these biomarkers predict the immune landscape in gastric adenocarcinoma-an aggressive cancer often linked to chronic inflammation-remains poorly understood. We leveraged high-dimensional spectral cytometry to generate a comprehensive single-cell immune landscape of tumors, normal tissue, and lymph nodes from patients in the Western Hemisphere with gastric adenocarcinoma. The immune composition of gastric tumors could not be predicted by traditional metrics such as tumor histology, molecular classification, mutational burden, or PD-L1 expression via IHC. Instead, our findings revealed that innate immune surveillance within tumors could be anticipated by the immune profile of the normal gastric mucosa. Additionally, distinct T-cell states in the lymph nodes were linked to the accumulation of activated and memory-like CD8+ tumor-infiltrating lymphocytes (TILs). Unbiased re-classification of patients based on tumor-specific immune infiltrate generated four distinct subtypes with varying immune compositions. Tumors with a T-cell-dominant immune subtype, which spanned TCGA molecular subtypes, were exclusively associated with superior responses to immunotherapy. Parallel analysis of metastatic gastric cancer patients treated with immune checkpoint blockade showed that patients who responded to immunotherapy had a pre-treatment tumor composition that corresponded to a T-cell-dominant immune subtype from our analysis. Taken together, this work identifies key host-specific factors associated with intratumoral immune composition in gastric cancer and offers an immunological classification system that can effectively identify patients likely to benefit from immune-based therapies.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":" ","pages":""},"PeriodicalIF":8.1,"publicationDate":"2025-01-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142945090","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cancer immunology research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1