Cancer immunology research最新文献

筛选
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
Cancer immunology research 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
{"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
Cancer immunology research 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
{"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
Cancer immunology research 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
{"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
Cancer immunology research Pub Date : 2024-09-03 DOI: 10.1158/2326-6066.CIR-24-0479
Kyoung Eun Lee, Marina Pasca di Magliano
{"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
Cancer immunology research 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
{"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
Cancer immunology research 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
{"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
Cancer immunology research 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
{"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
Cancer immunology research 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
{"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
Cancer immunology research 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
{"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
Cancer immunology research 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
{"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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
相关产品
×
本文献相关产品
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信