首页 > 最新文献

Cancer immunology research最新文献

英文 中文
Integrated germline and somatic features reveal divergent immune pathways driving response to immune checkpoint blockade 种系和体细胞综合特征揭示了驱动免疫检查点阻断反应的不同免疫途径
IF 10.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-10 DOI: 10.1158/2326-6066.cir-24-0164
Timothy J. Sears, Meghana S. Pagadala, Andrea Castro, Ko-Han Lee, JungHo Kong, Kairi Tanaka, Scott M. Lippman, Maurizio Zanetti, Hannah Carter
Immune Checkpoint Blockade (ICB) has revolutionized cancer treatment, however the mechanisms determining patient response remain poorly understood. Here, we used machine learning to predict ICB response from germline and somatic biomarkers and interpreted the learned model to uncover putative mechanisms driving superior outcomes. Patients with higher infiltration of T follicular helper cells had responses even in the presence of defects in the class-I Major Histocompatibility Complex (MHC-I). Further investigation uncovered different ICB responses in tumors when responses were reliant on MHC-I versus MHC-II neoantigens. Despite similar response rates, MHC-II reliant responses were associated with significantly longer durable clinical benefit (Discovery: Median OS=63.6 vs. 34.5 months P=0.0074; Validation: Median OS=37.5 vs. 33.1 months, P=0.040). Characteristics of the tumor immune microenvironment reflected MHC neoantigen reliance, and analysis of immune checkpoints revealed LAG3 as a potential target in MHC-II but not MHC-I reliant responses. This study highlights the value of interpretable machine learning models in elucidating the biological basis of therapy responses.
免疫检查点阻断疗法(ICB)给癌症治疗带来了革命性的变化,但人们对决定患者反应的机制仍然知之甚少。在这里,我们利用机器学习从种系和体细胞生物标志物中预测ICB反应,并对所学模型进行解释,以揭示驱动卓越疗效的假定机制。即使存在 I 类主要组织相容性复合物(MHC-I)缺陷,T 滤泡辅助细胞浸润较高的患者也能产生反应。进一步的研究发现,当肿瘤中的 ICB 反应依赖于 MHC-I 和 MHC-II 新抗原时,两者的反应有所不同。尽管反应率相似,但依赖 MHC-II 的反应与明显更长的持久临床获益相关(发现:中位 OS=63.6 个月 vs. 34.5 个月 P=0.0074;Validation:中位OS=37.5个月 vs. 33.1个月,P=0.040)。肿瘤免疫微环境的特征反映了对MHC新抗原的依赖性,免疫检查点分析显示LAG3是MHC-II而非MHC-I依赖性反应的潜在靶点。这项研究强调了可解释的机器学习模型在阐明治疗反应的生物学基础方面的价值。
{"title":"Integrated germline and somatic features reveal divergent immune pathways driving response to immune checkpoint blockade","authors":"Timothy J. Sears, Meghana S. Pagadala, Andrea Castro, Ko-Han Lee, JungHo Kong, Kairi Tanaka, Scott M. Lippman, Maurizio Zanetti, Hannah Carter","doi":"10.1158/2326-6066.cir-24-0164","DOIUrl":"https://doi.org/10.1158/2326-6066.cir-24-0164","url":null,"abstract":"Immune Checkpoint Blockade (ICB) has revolutionized cancer treatment, however the mechanisms determining patient response remain poorly understood. Here, we used machine learning to predict ICB response from germline and somatic biomarkers and interpreted the learned model to uncover putative mechanisms driving superior outcomes. Patients with higher infiltration of T follicular helper cells had responses even in the presence of defects in the class-I Major Histocompatibility Complex (MHC-I). Further investigation uncovered different ICB responses in tumors when responses were reliant on MHC-I versus MHC-II neoantigens. Despite similar response rates, MHC-II reliant responses were associated with significantly longer durable clinical benefit (Discovery: Median OS=63.6 vs. 34.5 months P=0.0074; Validation: Median OS=37.5 vs. 33.1 months, P=0.040). Characteristics of the tumor immune microenvironment reflected MHC neoantigen reliance, and analysis of immune checkpoints revealed LAG3 as a potential target in MHC-II but not MHC-I reliant responses. This study highlights the value of interpretable machine learning models in elucidating the biological basis of therapy responses.","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":10.1,"publicationDate":"2024-09-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142196793","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
Spatially resolved whole-transcriptomic and proteomic profiling of lung cancer and its immune-microenvironment according to PD-L1 expression. 根据 PD-L1 表达对肺癌及其免疫微环境进行空间解析的全转录组学和蛋白质组学分析。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-05 DOI: 10.1158/2326-6066.CIR-24-0071
Yoon Kyung Jeon, Jaemoon Koh, Dongjoo Lee, Sehui Kim, Seung Geun Song, Bogyeong Han, Hyein Jeong, Young A Kim, Bhumsuk Keam, Se-Hoon Lee, Kwangsoo Kim, Doo Hyun Chung

The expression of PD-L1 on tumor cells (TCs) is used as an immunotherapy biomarker in lung cancer, but heterogeneous intratumoral expression is often observed. Using a Digital Spatial Profiling, we performed proteomic and whole-transcriptomic analyses of TCs and immune cells (ICs) in spatially matched areas based on tumor PD-L1 expression and the status of the immune microenvironment. Our findings were validated using immunohistochemistry, The Cancer Genome Atlas, and immunotherapy cohorts. ICs in areas with high PD-L1 expression on TCs showed more features indicative of immunosuppression and exhaustion than ICs in areas with low PD-L1 expression on TCs. TCs highly expressing PD-L1 within immune-inflamed (IF) areas show up-regulation of pro-inflammatory processes, whereas TCs highly expressing PD-L1 within immune-deficient (ID) areas show up-regulation of various metabolic processes. Using differentially expressed genes of TCs between the IF and ID areas, we identified a novel prognostic gene signature for lung cancer. In addition, a high ratio of CD8+ cells to M2 macrophages was found to predict favorable outcomes in patients with PD-L1-expressing lung cancer after immune checkpoint inhibitor therapy. This study demonstrates that TCs and ICs have distinct spatial features within the tumor microenvironment that are related to tumor PD-L1 expression and IC infiltration.

肿瘤细胞(TC)上的PD-L1表达被用作肺癌的免疫治疗生物标记物,但经常观察到肿瘤内的异质性表达。我们利用数字空间剖析技术,根据肿瘤 PD-L1 的表达和免疫微环境的状况,对空间匹配区域的肿瘤细胞和免疫细胞(IC)进行了蛋白质组和全转录组分析。我们的研究结果得到了免疫组化、癌症基因组图谱和免疫疗法队列的验证。TC上PD-L1高表达区域的IC比TC上PD-L1低表达区域的IC显示出更多表明免疫抑制和衰竭的特征。免疫炎症(IF)区内高表达 PD-L1 的 TC 显示了促炎过程的上调,而免疫缺陷(ID)区内高表达 PD-L1 的 TC 则显示了各种代谢过程的上调。利用IF和ID区域TC的差异表达基因,我们发现了肺癌的新型预后基因特征。此外,我们还发现,CD8+细胞与M2巨噬细胞的高比例可预测PD-L1表达的肺癌患者在接受免疫检查点抑制剂治疗后的良好预后。这项研究表明,TC 和 IC 在肿瘤微环境中具有不同的空间特征,这些特征与肿瘤 PD-L1 表达和 IC 浸润有关。
{"title":"Spatially resolved whole-transcriptomic and proteomic profiling of lung cancer and its immune-microenvironment according to PD-L1 expression.","authors":"Yoon Kyung Jeon, Jaemoon Koh, Dongjoo Lee, Sehui Kim, Seung Geun Song, Bogyeong Han, Hyein Jeong, Young A Kim, Bhumsuk Keam, Se-Hoon Lee, Kwangsoo Kim, Doo Hyun Chung","doi":"10.1158/2326-6066.CIR-24-0071","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-24-0071","url":null,"abstract":"<p><p>The expression of PD-L1 on tumor cells (TCs) is used as an immunotherapy biomarker in lung cancer, but heterogeneous intratumoral expression is often observed. Using a Digital Spatial Profiling, we performed proteomic and whole-transcriptomic analyses of TCs and immune cells (ICs) in spatially matched areas based on tumor PD-L1 expression and the status of the immune microenvironment. Our findings were validated using immunohistochemistry, The Cancer Genome Atlas, and immunotherapy cohorts. ICs in areas with high PD-L1 expression on TCs showed more features indicative of immunosuppression and exhaustion than ICs in areas with low PD-L1 expression on TCs. TCs highly expressing PD-L1 within immune-inflamed (IF) areas show up-regulation of pro-inflammatory processes, whereas TCs highly expressing PD-L1 within immune-deficient (ID) areas show up-regulation of various metabolic processes. Using differentially expressed genes of TCs between the IF and ID areas, we identified a novel prognostic gene signature for lung cancer. In addition, a high ratio of CD8+ cells to M2 macrophages was found to predict favorable outcomes in patients with PD-L1-expressing lung cancer after immune checkpoint inhibitor therapy. This study demonstrates that TCs and ICs have distinct spatial features within the tumor microenvironment that are related to tumor PD-L1 expression and IC infiltration.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142131938","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of Core Techniques That Enhance Genome Editing of Human T Cells Expressing Synthetic Antigen Receptors. 确定可增强表达合成抗原受体的人类 T 细胞基因组编辑的核心技术。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-24-0251
Ju-Fang Chang, Nils Wellhausen, Nils W Engel, Jack H Landmann, Caitlin R Hopkins, January Salas-McKee, Adham S Bear, Mehmet E Selli, Sangya Agarwal, Julie K Jadlowsky, Gerald P Linette, Saar Gill, Carl H June, Joseph A Fraietta, Nathan Singh

Genome editing technologies have seen remarkable progress in recent years, enabling precise regulation of exogenous and endogenous genes. These advances have been extensively applied to the engineering of human T lymphocytes, leading to the development of practice changing therapies for patients with cancer and the promise of synthetic immune cell therapies for a variety of nonmalignant diseases. Many distinct conceptual and technical approaches have been used to edit T-cell genomes, however targeted assessments of which techniques are most effective for manufacturing, gene editing, and transgene expression are rarely reported. Through extensive comparative evaluation, we identified methods that most effectively enhance engineering of research-scale and preclinical T-cell products at critical stages of manufacturing.

近年来,基因组编辑技术取得了显著进展,实现了对外源和内源基因的精确调控。这些进展已被广泛应用于人类 T 淋巴细胞的工程设计,为癌症患者开发出了改变治疗方法的疗法,并有望为各种非恶性疾病开发出合成免疫细胞疗法。许多不同的概念和技术方法已被用于编辑 T 细胞基因组,但对哪些技术在制造、基因编辑和转基因表达方面最有效的针对性评估却鲜有报道。通过广泛的比较评估,我们确定了在制造的关键阶段最有效地提高研究规模和临床前 T 细胞产品工程的方法。
{"title":"Identification of Core Techniques That Enhance Genome Editing of Human T Cells Expressing Synthetic Antigen Receptors.","authors":"Ju-Fang Chang, Nils Wellhausen, Nils W Engel, Jack H Landmann, Caitlin R Hopkins, January Salas-McKee, Adham S Bear, Mehmet E Selli, Sangya Agarwal, Julie K Jadlowsky, Gerald P Linette, Saar Gill, Carl H June, Joseph A Fraietta, Nathan Singh","doi":"10.1158/2326-6066.CIR-24-0251","DOIUrl":"10.1158/2326-6066.CIR-24-0251","url":null,"abstract":"<p><p>Genome editing technologies have seen remarkable progress in recent years, enabling precise regulation of exogenous and endogenous genes. These advances have been extensively applied to the engineering of human T lymphocytes, leading to the development of practice changing therapies for patients with cancer and the promise of synthetic immune cell therapies for a variety of nonmalignant diseases. Many distinct conceptual and technical approaches have been used to edit T-cell genomes, however targeted assessments of which techniques are most effective for manufacturing, gene editing, and transgene expression are rarely reported. Through extensive comparative evaluation, we identified methods that most effectively enhance engineering of research-scale and preclinical T-cell products at critical stages of manufacturing.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141310129","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
Unveiling the Culprit: IL17 Signaling in the Pancreatic Epithelium Drives Tumorigenesis. 揭开罪魁祸首:胰腺上皮细胞中的 IL17 信号驱动肿瘤发生
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-24-0479
Kyoung Eun Lee, Marina Pasca di Magliano

IL17 signaling promotes pancreatic cancer development, yet the cell compartment responsible for the protumorigenic function of IL17 has not been defined. In this article, Castro-Pando and colleagues demonstrate that IL17/IL17 receptor A signaling in the pancreatic epithelium is critical for pancreatic cancer initiation and for establishing immunosuppression, whereas its signaling in the immune compartment is dispensable. This work provides an important mechanistic insight on the role of IL17 signaling and identifies a potential new immune checkpoint as a target in pancreatic cancer. See related article by Castro-Pando et al., p. 1170.

IL17 信号能促进胰腺癌的发展,但负责 IL17 原发肿瘤功能的细胞区系尚未确定。在这篇文章中,Castro-Pando 及其同事证明了胰腺上皮细胞中的 IL17/IL17 受体 A 信号传导对于胰腺癌的发生和免疫抑制的建立至关重要,而其在免疫细胞区的信号传导则是可有可无的。这项研究从机理上揭示了 IL17 信号传导的重要作用,并确定了一个潜在的新免疫检查点作为胰腺癌的靶点。参见 Castro-Pando 等人的相关文章,第 XXX (3) 页。
{"title":"Unveiling the Culprit: IL17 Signaling in the Pancreatic Epithelium Drives Tumorigenesis.","authors":"Kyoung Eun Lee, Marina Pasca di Magliano","doi":"10.1158/2326-6066.CIR-24-0479","DOIUrl":"10.1158/2326-6066.CIR-24-0479","url":null,"abstract":"<p><p>IL17 signaling promotes pancreatic cancer development, yet the cell compartment responsible for the protumorigenic function of IL17 has not been defined. In this article, Castro-Pando and colleagues demonstrate that IL17/IL17 receptor A signaling in the pancreatic epithelium is critical for pancreatic cancer initiation and for establishing immunosuppression, whereas its signaling in the immune compartment is dispensable. This work provides an important mechanistic insight on the role of IL17 signaling and identifies a potential new immune checkpoint as a target in pancreatic cancer. See related article by Castro-Pando et al., p. 1170.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141579028","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
Intracellular Osteopontin Promotes the Release of TNFα by Mast Cells to Restrain Neuroendocrine Prostate Cancer. 细胞内补骨脂素促进肥大细胞释放 TNF 以抑制神经内分泌性前列腺癌。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-23-0792
Roberta Sulsenti, Giuseppina B Scialpi, Barbara Frossi, Laura Botti, Renata Ferri, Irene Tripodi, Annamaria Piva, Sabina Sangaletti, Davide Pernici, Valeria Cancila, Francesco Romeo, Claudia Chiodoni, Daniele Lecis, Francesca Bianchi, Irene Fischetti, Claudia Enriquez, Filippo Crivelli, Marco Bregni, Giuseppe Renne, Salvatore Pece, Claudio Tripodo, Carlo E Pucillo, Mario P Colombo, Elena Jachetti

Neuroendocrine prostate cancer (NEPC) is an aggressive form of prostate cancer that emerges as tumors become resistant to hormone therapies or, rarely, arises de novo in treatment-naïve patients. The urgent need for effective therapies against NEPC is hampered by the limited knowledge of the biology governing this lethal disease. Based on our prior observations in the transgenic adenocarcinoma of the mouse prostate (TRAMP) spontaneous prostate cancer model, in which the genetic depletion of either mast cells (MC) or the matricellular protein osteopontin (OPN) increases NEPC frequency, we tested the hypothesis that MCs can restrain NEPC through OPN production, using in vitro co-cultures between murine or human tumor cell lines and MCs, and in vivo experiments. We unveiled a role for the intracellular isoform of OPN, so far neglected compared with the secreted isoform. Mechanistically, we unraveled that the intracellular isoform of OPN promotes TNFα production in MCs via the TLR2/TLR4-MyD88 axis, specifically triggered by the encounter with NEPC cells. We found that MC-derived TNFα, in turn, hampered the growth of NEPC. We then identified the protein syndecan-1 (SDC1) as the NEPC-specific TLR2/TLR4 ligand that triggered this pathway. Interrogating published single-cell RNA-sequencing data, we validated this mechanism in a different mouse model. Translational relevance of the results was provided by in silico analyses of available human NEPC datasets and by immunofluorescence on patient-derived adenocarcinoma and NEPC lesions. Overall, our results show that MCs actively inhibit NEPC, paving the way for innovative MC-based therapies for this fatal tumor. We also highlight SDC1 as a potential biomarker for incipient NEPC.

神经内分泌性前列腺癌(NEPC)是一种侵袭性前列腺癌,当肿瘤对激素疗法产生耐药性时就会出现,或者在极少数情况下,在治疗无效的患者中从头开始出现。由于对这种致命疾病的生物学特性了解有限,因此迫切需要针对 NEPC 的有效疗法。在TRAMP自发性前列腺癌模型中,肥大细胞(MCs)或基质蛋白骨通素(OPN)的基因消耗会增加NEPC的发生频率,基于之前的观察结果,我们利用小鼠或人类肿瘤细胞系与MCs的体外共培养以及体内实验,验证了MCs能通过产生OPN抑制NEPC的假设。我们揭示了细胞内异构体 OPN(iOPN)的作用,与分泌型异构体相比,细胞内异构体的作用至今仍被忽视。从机理上讲,我们揭示了iOPN通过TLR2/TLR4-MyD88轴促进MCs产生TNF,特别是在与NEPC细胞相遇时触发。我们发现,MC衍生的TNF,反过来又阻碍了NEPC的生长。然后,我们发现蛋白辛迪加-1(SDC1)是触发这一途径的NEPC特异性TLR2/TLR4配体。通过研究已发表的单细胞 RNA 序列数据,我们在不同的小鼠模型中验证了这一机制。通过对现有的人类 NEPC 数据集进行默片分析,以及对源自患者的腺癌和 NEPC 病变进行免疫荧光,证明了这些结果的转化相关性。总之,我们的研究结果表明 MCs 能积极抑制 NEPC,为基于 MCs 的创新疗法治疗这种致命肿瘤铺平了道路。我们还强调了 SDC1 作为萌芽期 NEPC 潜在生物标志物的作用。
{"title":"Intracellular Osteopontin Promotes the Release of TNFα by Mast Cells to Restrain Neuroendocrine Prostate Cancer.","authors":"Roberta Sulsenti, Giuseppina B Scialpi, Barbara Frossi, Laura Botti, Renata Ferri, Irene Tripodi, Annamaria Piva, Sabina Sangaletti, Davide Pernici, Valeria Cancila, Francesco Romeo, Claudia Chiodoni, Daniele Lecis, Francesca Bianchi, Irene Fischetti, Claudia Enriquez, Filippo Crivelli, Marco Bregni, Giuseppe Renne, Salvatore Pece, Claudio Tripodo, Carlo E Pucillo, Mario P Colombo, Elena Jachetti","doi":"10.1158/2326-6066.CIR-23-0792","DOIUrl":"10.1158/2326-6066.CIR-23-0792","url":null,"abstract":"<p><p>Neuroendocrine prostate cancer (NEPC) is an aggressive form of prostate cancer that emerges as tumors become resistant to hormone therapies or, rarely, arises de novo in treatment-naïve patients. The urgent need for effective therapies against NEPC is hampered by the limited knowledge of the biology governing this lethal disease. Based on our prior observations in the transgenic adenocarcinoma of the mouse prostate (TRAMP) spontaneous prostate cancer model, in which the genetic depletion of either mast cells (MC) or the matricellular protein osteopontin (OPN) increases NEPC frequency, we tested the hypothesis that MCs can restrain NEPC through OPN production, using in vitro co-cultures between murine or human tumor cell lines and MCs, and in vivo experiments. We unveiled a role for the intracellular isoform of OPN, so far neglected compared with the secreted isoform. Mechanistically, we unraveled that the intracellular isoform of OPN promotes TNFα production in MCs via the TLR2/TLR4-MyD88 axis, specifically triggered by the encounter with NEPC cells. We found that MC-derived TNFα, in turn, hampered the growth of NEPC. We then identified the protein syndecan-1 (SDC1) as the NEPC-specific TLR2/TLR4 ligand that triggered this pathway. Interrogating published single-cell RNA-sequencing data, we validated this mechanism in a different mouse model. Translational relevance of the results was provided by in silico analyses of available human NEPC datasets and by immunofluorescence on patient-derived adenocarcinoma and NEPC lesions. Overall, our results show that MCs actively inhibit NEPC, paving the way for innovative MC-based therapies for this fatal tumor. We also highlight SDC1 as a potential biomarker for incipient NEPC.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11369624/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141310130","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
Endoplasmic Reticulum Stress Potentiates the Immunosuppressive Microenvironment in Hepatocellular Carcinoma by Promoting the Release of SNHG6-Enriched Small Extracellular Vesicles. 内质网应激通过促进富含 SNHG6 的小细胞外囊泡的释放,增强肝细胞癌的免疫抑制微环境。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-23-0469
Chengdong Liu, Xiaohan Zhou, Hanyi Zeng, Jiaping Yu, Wenwen Li, Wanli Zhang, Yanxia Liao, Haijian Wang, Li Liu

Endoplasmic reticulum (ER) stress leads to hepatocellular carcinoma (HCC) progression. Small extracellular vesicles (sEV) play a crucial role in modulating the tumor microenvironment (TME) by influencing cellular communication and immune responses. However, it is unclear whether ER stress modulates the TME through sEVs. In the current study, we investigated the effects and underlying mechanisms of ER stress on the HCC TME. In vivo and in vitro experiments showed that overactivated ER stress was a salient attribute of the immunosuppressive HCC TME. This was caused by the ATF4-promoted release of small nucleolar RNA host gene 6 (SNHG6)-carrying sEVs, which attenuated T cell-mediated immune responses. Overall, SNHG6 modulated the immunosuppressive TME and aggravated ER stress. Meanwhile, targeting SNHG6 facilitated M1-like macrophage and CD8+ T-cell infiltration and decreased the proportion of M2-like macrophages. In addition, SNHG6 knockdown enhanced anti-PD1 immunotherapeutic efficacy. Moreover, in HCC patients, overexpression of SNHG6 was associated with a lack of response to anti-PD1 therapy and poor prognosis, whereas low SNHG6 expression was associated with improved therapeutic efficacy and prognoses. These data indicate that a correlation exists among ER stress, sEVs, immunosuppressive HCC TME, and immunotherapeutic efficacy. Hence, SNHG6-targeted therapy may represent an effective strategy for patients with HCC.

内质网(ER)应激会导致肝细胞癌(HCC)恶化。小细胞外囊泡(sEVs)通过影响细胞通讯和免疫反应,在调节肿瘤微环境(TME)方面发挥着至关重要的作用。然而,目前还不清楚ER应激是否会通过sEVs调节肿瘤微环境。在本研究中,我们探讨了ER应激对HCC TME的影响及其内在机制。体内和体外实验表明,过度激活的ER应激是免疫抑制性HCC TME的一个显著特征。这是由于 ATF4 促进了携带 SNHG6 的 sEVs 的释放,从而削弱了 T 细胞介导的免疫反应。总之,SNHG6调节了免疫抑制性TME并加重了ER应激。同时,靶向 SNHG6 可促进 M1 样巨噬细胞和 CD8+ T 细胞浸润,并降低 M2 样巨噬细胞的比例。此外,敲除SNHG6还能增强抗PD-1免疫治疗的疗效。此外,在 HCC 患者中,SNHG6 的过表达与抗 PD-1 治疗缺乏反应和预后不良有关,而 SNHG6 的低表达与治疗效果和预后的改善有关。这些数据表明,ER 应激、sEVs、免疫抑制性 HCC TME 和免疫治疗效果之间存在相关性。因此,SNHG6靶向治疗可能是治疗HCC患者的有效策略。
{"title":"Endoplasmic Reticulum Stress Potentiates the Immunosuppressive Microenvironment in Hepatocellular Carcinoma by Promoting the Release of SNHG6-Enriched Small Extracellular Vesicles.","authors":"Chengdong Liu, Xiaohan Zhou, Hanyi Zeng, Jiaping Yu, Wenwen Li, Wanli Zhang, Yanxia Liao, Haijian Wang, Li Liu","doi":"10.1158/2326-6066.CIR-23-0469","DOIUrl":"10.1158/2326-6066.CIR-23-0469","url":null,"abstract":"<p><p>Endoplasmic reticulum (ER) stress leads to hepatocellular carcinoma (HCC) progression. Small extracellular vesicles (sEV) play a crucial role in modulating the tumor microenvironment (TME) by influencing cellular communication and immune responses. However, it is unclear whether ER stress modulates the TME through sEVs. In the current study, we investigated the effects and underlying mechanisms of ER stress on the HCC TME. In vivo and in vitro experiments showed that overactivated ER stress was a salient attribute of the immunosuppressive HCC TME. This was caused by the ATF4-promoted release of small nucleolar RNA host gene 6 (SNHG6)-carrying sEVs, which attenuated T cell-mediated immune responses. Overall, SNHG6 modulated the immunosuppressive TME and aggravated ER stress. Meanwhile, targeting SNHG6 facilitated M1-like macrophage and CD8+ T-cell infiltration and decreased the proportion of M2-like macrophages. In addition, SNHG6 knockdown enhanced anti-PD1 immunotherapeutic efficacy. Moreover, in HCC patients, overexpression of SNHG6 was associated with a lack of response to anti-PD1 therapy and poor prognosis, whereas low SNHG6 expression was associated with improved therapeutic efficacy and prognoses. These data indicate that a correlation exists among ER stress, sEVs, immunosuppressive HCC TME, and immunotherapeutic efficacy. Hence, SNHG6-targeted therapy may represent an effective strategy for patients with HCC.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141426398","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
Cannabidiol enhances Atezolizumab efficacy by upregulating PD-L1 expression via the cGAS-STING pathway in triple-negative breast cancer cells. 大麻二酚通过 cGAS-STING 通路上调三阴性乳腺癌细胞中 PD-L1 的表达,从而增强阿特珠单抗的疗效。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-23-0902
Bu Gyeom Kim, Bo Ram Kim, Dae Yeong Kim, Woo Young Kim, Sanghee Kang, Sun Il Lee, Sang Cheul Oh

The treatment of patients with triple negative breast cancer (TNBC) relies on cytotoxic therapy. Currently, atezolizumab and chemotherapy can be combined in patients with TNBC. However, this approach is not effective for all patients with low reactivity to atezolizumab. As there is a lack of alternative treatment options, new anti-cancer drugs are urgently needed to enhance atezolizumab reactivity against TNBC. Recent strategies have focused on regulating the expression of programmed death-ligand 1 (PD-L1) or enhancing immune response activation by combining anti-cancer drugs with immune checkpoint inhibitors (ICIs). Cannabidiol (CBD), a cannabinoid component derived from the cannabis plant, has been reported to have anti-cancer therapeutic potential because of its capacity to induce apoptotic cell death in tumor cells while avoiding cytotoxicity in normal cells. Previous studies have demonstrated the effects of CBD on apoptosis in various cancer cell types. However, the potential role of CBD as an immune modulator in the regulation of PD-L1 expression and anti-cancer immune responses remains to be explored. In this study, we found that CBD stimulated PD-L1 expression in TNBC cells, which significantly induced the CBD-mediated cGAS-STING pathway activation. Taken together, we demonstrated that the combination of CBD and anti-PD-L1 antibody enhances the anti-cancer immune response in vitro and in vivo experiments. Our findings identified the mechanism of PD-L1 regulation by CBD in TNBC cells and suggested that CBD could be a potential candidate for the development of new combinatorial strategies with ICIs in TNBC patients.

三阴性乳腺癌(TNBC)患者的治疗主要依靠细胞毒疗法。目前,atezolizumab 和化疗可联合用于 TNBC 患者。然而,这种方法并不适用于所有对 atezolizumab 反应性低的患者。由于缺乏替代治疗方案,因此迫切需要新的抗癌药物来提高atezolizumab对TNBC的反应性。近期的策略主要集中在调节程序性死亡配体1(PD-L1)的表达,或通过将抗癌药物与免疫检查点抑制剂(ICIs)相结合来增强免疫反应的激活。据报道,大麻二酚(CBD)是从大麻植物中提取的一种大麻素成分,具有抗癌治疗潜力,因为它能诱导肿瘤细胞凋亡,同时避免对正常细胞产生细胞毒性。以前的研究已经证明了 CBD 对各种癌症细胞凋亡的影响。然而,CBD 作为免疫调节剂在调节 PD-L1 表达和抗癌免疫反应方面的潜在作用仍有待探索。在本研究中,我们发现 CBD 可刺激 TNBC 细胞中 PD-L1 的表达,从而显著诱导 CBD 介导的 cGAS-STING 通路活化。综上所述,我们证明了 CBD 和抗 PD-L1 抗体的联合使用可增强体外和体内实验中的抗癌免疫反应。我们的研究结果确定了CBD在TNBC细胞中调控PD-L1的机制,并表明CBD可能是TNBC患者与ICIs联合开发新策略的潜在候选药物。
{"title":"Cannabidiol enhances Atezolizumab efficacy by upregulating PD-L1 expression via the cGAS-STING pathway in triple-negative breast cancer cells.","authors":"Bu Gyeom Kim, Bo Ram Kim, Dae Yeong Kim, Woo Young Kim, Sanghee Kang, Sun Il Lee, Sang Cheul Oh","doi":"10.1158/2326-6066.CIR-23-0902","DOIUrl":"https://doi.org/10.1158/2326-6066.CIR-23-0902","url":null,"abstract":"<p><p>The treatment of patients with triple negative breast cancer (TNBC) relies on cytotoxic therapy. Currently, atezolizumab and chemotherapy can be combined in patients with TNBC. However, this approach is not effective for all patients with low reactivity to atezolizumab. As there is a lack of alternative treatment options, new anti-cancer drugs are urgently needed to enhance atezolizumab reactivity against TNBC. Recent strategies have focused on regulating the expression of programmed death-ligand 1 (PD-L1) or enhancing immune response activation by combining anti-cancer drugs with immune checkpoint inhibitors (ICIs). Cannabidiol (CBD), a cannabinoid component derived from the cannabis plant, has been reported to have anti-cancer therapeutic potential because of its capacity to induce apoptotic cell death in tumor cells while avoiding cytotoxicity in normal cells. Previous studies have demonstrated the effects of CBD on apoptosis in various cancer cell types. However, the potential role of CBD as an immune modulator in the regulation of PD-L1 expression and anti-cancer immune responses remains to be explored. In this study, we found that CBD stimulated PD-L1 expression in TNBC cells, which significantly induced the CBD-mediated cGAS-STING pathway activation. Taken together, we demonstrated that the combination of CBD and anti-PD-L1 antibody enhances the anti-cancer immune response in vitro and in vivo experiments. Our findings identified the mechanism of PD-L1 regulation by CBD in TNBC cells and suggested that CBD could be a potential candidate for the development of new combinatorial strategies with ICIs in TNBC patients.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142124909","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
MerTK Induces Dysfunctional Dendritic Cells by Metabolic Reprogramming. MerTK 通过代谢重编程诱导功能失调的树突状细胞
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-23-0666
Eden Y Zewdie, George M Edwards, Debra M Hunter, Henry Shelton Earp, Alisha Holtzhausen

Checkpoint inhibitors, specifically anti-programmed cell death protein 1 (PD1), have shown success in treating metastatic melanoma; however, some patients develop resistance. Dendritic cells (DC) play a key role in initiating an immune response, but in certain circumstances they become ineffective. We investigated the role of MerTK, a receptor tyrosine kinase responsible for myeloid cell clearance of dead cells, in the regulation of DC function and metabolism in the tumor microenvironment. Tumors resistant to anti-PD1 exhibited increased levels of MerTK+ DCs. Treating wild-type DCs with apoptotic melanoma cells in vitro resulted in increased MerTK expression, elevated mitochondrial respiration and fatty acid oxidation, and reduced T-cell stimulatory capacity, all characteristics of dysfunctional DCs. In contrast, dead cells had only limited effect on the metabolism of MerTK-deficient DCs, which instead maintained an antigen-presenting, stimulatory phenotype. The efficacy of anti-PD1 to slow tumor progression and induce antigen specific T-cell infiltration was markedly increased in mice with selective ablation of MerTK in the DC compartment, suggesting the possibility of therapeutically targeting MerTK to modulate DC metabolism and function and enhance anti-PD1 therapy.

检查点抑制剂,特别是抗-PD-1,在治疗转移性黑色素瘤方面取得了成功;但是,有些患者会产生抗药性。树突状细胞(DC)在启动免疫反应中发挥着关键作用,但在某些情况下它们会变得无效。我们研究了MerTK(一种负责髓系细胞清除死亡细胞的受体酪氨酸激酶)在肿瘤微环境中调控树突状细胞功能和新陈代谢中的作用。对抗PD-1耐药的肿瘤显示出MerTK+ DCs水平的升高。在体外用凋亡的黑色素瘤死细胞处理野生型直流细胞会导致 MerTK 表达增加、线粒体呼吸和脂肪酸氧化升高以及 T 细胞刺激能力降低,这些都是功能失调型直流细胞的特征。相比之下,死亡细胞对缺失 MerTK 的直流细胞的新陈代谢影响有限,相反,直流细胞保持了抗原呈递和刺激表型。在选择性消减直流区MerTK的小鼠中,抗PD-1减缓肿瘤进展和诱导特异性T细胞浸润的效果明显增强,这表明有可能以MerTK为治疗靶点,调节直流代谢和功能,加强抗PD-1治疗。
{"title":"MerTK Induces Dysfunctional Dendritic Cells by Metabolic Reprogramming.","authors":"Eden Y Zewdie, George M Edwards, Debra M Hunter, Henry Shelton Earp, Alisha Holtzhausen","doi":"10.1158/2326-6066.CIR-23-0666","DOIUrl":"10.1158/2326-6066.CIR-23-0666","url":null,"abstract":"<p><p>Checkpoint inhibitors, specifically anti-programmed cell death protein 1 (PD1), have shown success in treating metastatic melanoma; however, some patients develop resistance. Dendritic cells (DC) play a key role in initiating an immune response, but in certain circumstances they become ineffective. We investigated the role of MerTK, a receptor tyrosine kinase responsible for myeloid cell clearance of dead cells, in the regulation of DC function and metabolism in the tumor microenvironment. Tumors resistant to anti-PD1 exhibited increased levels of MerTK+ DCs. Treating wild-type DCs with apoptotic melanoma cells in vitro resulted in increased MerTK expression, elevated mitochondrial respiration and fatty acid oxidation, and reduced T-cell stimulatory capacity, all characteristics of dysfunctional DCs. In contrast, dead cells had only limited effect on the metabolism of MerTK-deficient DCs, which instead maintained an antigen-presenting, stimulatory phenotype. The efficacy of anti-PD1 to slow tumor progression and induce antigen specific T-cell infiltration was markedly increased in mice with selective ablation of MerTK in the DC compartment, suggesting the possibility of therapeutically targeting MerTK to modulate DC metabolism and function and enhance anti-PD1 therapy.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11371516/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141558156","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
PlexinB1 Inactivation Reprograms Immune Cells in the Tumor Microenvironment, Inhibiting Breast Cancer Growth and Metastatic Dissemination. PlexinB1 失活可对肿瘤微环境中的免疫细胞进行重编程,从而抑制乳腺癌的生长和转移扩散。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-23-0289
Giulia Franzolin, Serena Brundu, Carina F Cojocaru, Aurora Curatolo, Matteo Ponzo, Roberta Mastrantonio, Emiko Mihara, Atsushi Kumanogoh, Hiroaki Suga, Junichi Takagi, Luca Tamagnone, Enrico Giraudo

Semaphorin-plexin signaling plays a major role in the tumor microenvironment (TME). In particular, Semaphorin 4D (SEMA4D) has been shown to promote tumor growth and metastasis; however, the role of its high-affinity receptor Plexin-B1 (PLXNB1), which is expressed in the TME, is poorly understood. In this study, we directly targeted PLXNB1 in the TME of triple-negative murine breast carcinoma to elucidate its relevance in cancer progression. We found that primary tumor growth and metastatic dissemination were strongly reduced in PLXNB1-deficient mice, which showed longer survival. PLXNB1 loss in the TME induced a switch in the polarization of tumor-associated macrophages (TAM) toward a pro-inflammatory M1 phenotype and enhanced the infiltration of CD8+ T lymphocytes both in primary tumors and in distant metastases. Moreover, PLXNB1 deficiency promoted a shift in the Th1/Th2 balance of the T-cell population and an antitumor gene signature, with the upregulation of Icos, Perforin-1, Stat3, and Ccl5 in tumor-infiltrating lymphocytes (TILs). We thus tested the translational relevance of TME reprogramming driven by PLXNB1 inactivation for responsiveness to immunotherapy. Indeed, in the absence of PLXNB1, the efficacy of anti-PD-1 blockade was strongly enhanced, efficiently reducing tumor growth and distant metastasis. Consistent with this, pharmacological PLXNB1 blockade by systemic treatment with a specific inhibitor significantly hampered breast cancer growth and enhanced the antitumor activity of the anti-PD-1 treatment in a preclinical model. Altogether, these data indicate that PLXNB1 signaling controls the antitumor immune response in the TME and highlight this receptor as a promising immune therapeutic target for metastatic breast cancers.

Semaphorin-Plexin 信号在肿瘤微环境(TME)中发挥着重要作用。特别是,Semaphorin 4D (SEMA4D)已被证明能促进肿瘤生长和转移;然而,人们对其高亲和力受体 Plexin-B1 (PLXNB1)在肿瘤微环境中表达的作用却知之甚少。在本研究中,我们直接靶向三阴性小鼠乳腺癌TME中的PLXNB1,以阐明其在癌症进展中的相关性。我们发现,PLXNB1缺失的小鼠原发性肿瘤生长和转移扩散均显著减少,生存期也更长。TME中PLXNB1的缺失诱导肿瘤相关巨噬细胞(TAMs)向促炎M1表型极化,并增强了CD8+ T淋巴细胞在原发性肿瘤和远处转移灶中的浸润。此外,PLXNB1缺陷还促进了T细胞群Th1/Th2平衡的转变和抗肿瘤基因特征的形成,肿瘤浸润淋巴细胞(TILs)中的Icos、Perforin-1、Stat3和Ccl5均上调。因此,我们测试了由 PLXNB1 失活驱动的 TME 重编程对免疫疗法反应性的转化相关性。事实上,在 PLXNB1 缺失的情况下,抗 PD-1 阻断剂的疗效得到了显著增强,有效减少了肿瘤生长和远处转移。与此相一致的是,在临床前模型中,通过使用特异性抑制剂进行全身治疗对 PLXNB1 进行药理阻断,可显著抑制乳腺癌的生长,并增强抗 PD1 治疗的抗肿瘤活性。总之,这些数据表明 PLXNB1 信号传导控制着 TME 中的抗肿瘤免疫反应,并强调该受体是治疗转移性乳腺癌的一个有前景的免疫治疗靶点。
{"title":"PlexinB1 Inactivation Reprograms Immune Cells in the Tumor Microenvironment, Inhibiting Breast Cancer Growth and Metastatic Dissemination.","authors":"Giulia Franzolin, Serena Brundu, Carina F Cojocaru, Aurora Curatolo, Matteo Ponzo, Roberta Mastrantonio, Emiko Mihara, Atsushi Kumanogoh, Hiroaki Suga, Junichi Takagi, Luca Tamagnone, Enrico Giraudo","doi":"10.1158/2326-6066.CIR-23-0289","DOIUrl":"10.1158/2326-6066.CIR-23-0289","url":null,"abstract":"<p><p>Semaphorin-plexin signaling plays a major role in the tumor microenvironment (TME). In particular, Semaphorin 4D (SEMA4D) has been shown to promote tumor growth and metastasis; however, the role of its high-affinity receptor Plexin-B1 (PLXNB1), which is expressed in the TME, is poorly understood. In this study, we directly targeted PLXNB1 in the TME of triple-negative murine breast carcinoma to elucidate its relevance in cancer progression. We found that primary tumor growth and metastatic dissemination were strongly reduced in PLXNB1-deficient mice, which showed longer survival. PLXNB1 loss in the TME induced a switch in the polarization of tumor-associated macrophages (TAM) toward a pro-inflammatory M1 phenotype and enhanced the infiltration of CD8+ T lymphocytes both in primary tumors and in distant metastases. Moreover, PLXNB1 deficiency promoted a shift in the Th1/Th2 balance of the T-cell population and an antitumor gene signature, with the upregulation of Icos, Perforin-1, Stat3, and Ccl5 in tumor-infiltrating lymphocytes (TILs). We thus tested the translational relevance of TME reprogramming driven by PLXNB1 inactivation for responsiveness to immunotherapy. Indeed, in the absence of PLXNB1, the efficacy of anti-PD-1 blockade was strongly enhanced, efficiently reducing tumor growth and distant metastasis. Consistent with this, pharmacological PLXNB1 blockade by systemic treatment with a specific inhibitor significantly hampered breast cancer growth and enhanced the antitumor activity of the anti-PD-1 treatment in a preclinical model. Altogether, these data indicate that PLXNB1 signaling controls the antitumor immune response in the TME and highlight this receptor as a promising immune therapeutic target for metastatic breast cancers.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11369622/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141316753","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
Regulatory Considerations for Genome-Edited T-cell Therapies. 基因组编辑 T 细胞疗法的监管考虑因素。
IF 8.1 1区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-24-0482
Julie K Jadlowsky, Ju-Fang Chang, David H Spencer, John M Warrington, Bruce L Levine, Carl H June, Joseph A Fraietta, Nathan Singh

Methods to engineer the genomes of human cells for therapeutic intervention continue to advance at a remarkable pace. Chimeric antigen receptor-engineered T lymphocytes have pioneered the way for these therapies, initially beginning with insertions of chimeric antigen receptor transgenes into T-cell genomes using classical gene therapy vectors. The broad use of clustered regularly interspaced short palindromic repeats (CRISPR)-based technologies to edit endogenous genes has now opened the door to a new era of precision medicine. To add complexity, many engineered cellular therapies under development integrate gene therapy with genome editing to introduce novel biological functions and enhance therapeutic efficacy. Here, we review the current state of scientific, translational, and regulatory oversight of gene-edited cell products.

改造人体细胞基因组以进行治疗干预的方法正以惊人的速度不断进步。嵌合抗原受体工程 T 淋巴细胞开创了这些疗法的先河,最初是利用传统的基因治疗载体将嵌合抗原受体转基因插入 T 细胞基因组。现在,基于聚类规则间隔短回文重复序列(CRISPR)技术的广泛应用为编辑内源基因打开了一扇门,开启了精准医疗的新时代。为了增加复杂性,许多正在开发的工程细胞疗法将基因治疗与基因组编辑相结合,以引入新的生物功能并提高疗效。在此,我们回顾了基因编辑细胞产品的科学、转化和监管现状。
{"title":"Regulatory Considerations for Genome-Edited T-cell Therapies.","authors":"Julie K Jadlowsky, Ju-Fang Chang, David H Spencer, John M Warrington, Bruce L Levine, Carl H June, Joseph A Fraietta, Nathan Singh","doi":"10.1158/2326-6066.CIR-24-0482","DOIUrl":"10.1158/2326-6066.CIR-24-0482","url":null,"abstract":"<p><p>Methods to engineer the genomes of human cells for therapeutic intervention continue to advance at a remarkable pace. Chimeric antigen receptor-engineered T lymphocytes have pioneered the way for these therapies, initially beginning with insertions of chimeric antigen receptor transgenes into T-cell genomes using classical gene therapy vectors. The broad use of clustered regularly interspaced short palindromic repeats (CRISPR)-based technologies to edit endogenous genes has now opened the door to a new era of precision medicine. To add complexity, many engineered cellular therapies under development integrate gene therapy with genome editing to introduce novel biological functions and enhance therapeutic efficacy. Here, we review the current state of scientific, translational, and regulatory oversight of gene-edited cell products.</p>","PeriodicalId":9474,"journal":{"name":"Cancer immunology research","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11371504/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141626088","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
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