首页 > 最新文献

Oncoimmunology最新文献

英文 中文
High-grade serous ovarian cancer development and anti-PD-1 resistance is driven by IRE1α activity in neutrophils. 中性粒细胞中的IRE1α活性驱动了高级别浆液性卵巢癌的发展和抗PD-1的耐药性。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-02 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2411070
Alexander Emmanuelli, Camilla Salvagno, Sung-Min Hwang, Deepika Awasthi, Tito A Sandoval, Chang-Suk Chae, Jin-Gyu Cheong, Chen Tan, Takao Iwawaki, Juan R Cubillos-Ruiz

High-grade serious ovarian cancer (HGSOC) is an aggressive malignancy that remains refractory to current immunotherapies. While advanced stage disease has been extensively studied, the cellular and molecular mechanisms that promote early immune escape in HGSOC remain largely unexplored. Here, we report that primary HGSO tumors program neutrophils to inhibit T cell anti-tumor function by activating the endoplasmic reticulum (ER) stress sensor IRE1α. We found that intratumoral neutrophils exhibited overactivation of ER stress response markers compared with their counterparts at non-tumor sites. Selective deletion of IRE1α in neutrophils delayed primary ovarian tumor growth and extended the survival of mice with HGSOC by enabling early T cell-mediated tumor control. Notably, loss of IRE1α in neutrophils sensitized tumor-bearing mice to PD-1 blockade, inducing HGSOC regression and long-term survival in ~ 50% of the treated hosts. Hence, neutrophil-intrinsic IRE1α facilitates early adaptive immune escape in HGSOC and targeting this ER stress sensor might be used to unleash endogenous and immunotherapy-elicited immunity that controls metastatic disease.

高级别重度卵巢癌(HGSOC)是一种侵袭性恶性肿瘤,对目前的免疫疗法仍有耐药性。虽然晚期疾病已被广泛研究,但促进 HGSOC 早期免疫逃逸的细胞和分子机制在很大程度上仍未被探索。在这里,我们报告了原发性HGSO肿瘤通过激活内质网(ER)应激传感器IRE1α,使中性粒细胞抑制T细胞的抗肿瘤功能。我们发现,与非肿瘤部位的中性粒细胞相比,瘤内中性粒细胞表现出过度激活ER应激反应标记物。选择性地删除中性粒细胞中的IRE1α可延缓原发性卵巢肿瘤的生长,并通过早期T细胞介导的肿瘤控制延长HGSOC小鼠的生存期。值得注意的是,中性粒细胞中 IRE1α 的缺失会使肿瘤小鼠对 PD-1 阻滞剂敏感,从而诱导 HGSOC 消退,并使约 50% 的受试宿主长期存活。因此,中性粒细胞内在的IRE1α有助于HGSOC早期适应性免疫逃逸,靶向这种ER应激传感器可用于释放内源性免疫和免疫疗法诱导的免疫,从而控制转移性疾病。
{"title":"High-grade serous ovarian cancer development and anti-PD-1 resistance is driven by IRE1α activity in neutrophils.","authors":"Alexander Emmanuelli, Camilla Salvagno, Sung-Min Hwang, Deepika Awasthi, Tito A Sandoval, Chang-Suk Chae, Jin-Gyu Cheong, Chen Tan, Takao Iwawaki, Juan R Cubillos-Ruiz","doi":"10.1080/2162402X.2024.2411070","DOIUrl":"10.1080/2162402X.2024.2411070","url":null,"abstract":"<p><p>High-grade serious ovarian cancer (HGSOC) is an aggressive malignancy that remains refractory to current immunotherapies. While advanced stage disease has been extensively studied, the cellular and molecular mechanisms that promote early immune escape in HGSOC remain largely unexplored. Here, we report that primary HGSO tumors program neutrophils to inhibit T cell anti-tumor function by activating the endoplasmic reticulum (ER) stress sensor IRE1α. We found that intratumoral neutrophils exhibited overactivation of ER stress response markers compared with their counterparts at non-tumor sites. Selective deletion of IRE1α in neutrophils delayed primary ovarian tumor growth and extended the survival of mice with HGSOC by enabling early T cell-mediated tumor control. Notably, loss of IRE1α in neutrophils sensitized tumor-bearing mice to PD-1 blockade, inducing HGSOC regression and long-term survival in ~ 50% of the treated hosts. Hence, neutrophil-intrinsic IRE1α facilitates early adaptive immune escape in HGSOC and targeting this ER stress sensor might be used to unleash endogenous and immunotherapy-elicited immunity that controls metastatic disease.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448341/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142373288","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Clinical prognosticators and targets in the immune microenvironment of intrahepatic cholangiocarcinoma. 肝内胆管癌免疫微环境中的临床预后指标和靶点。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-10-01 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2406052
Isis Lozzi, Alexander Arnold, Matthias Barone, Juliette Claire Johnson, Bruno V Sinn, Johannes Eschrich, Pimrapat Gebert, Ruonan Wang, Mengwen Hu, Linda Feldbrügge, Anja Schirmeier, Anja Reutzel-Selke, Thomas Malinka, Felix Krenzien, Wenzel Schöning, Dominik P Modest, Johann Pratschke, Igor M Sauer, Matthäus Felsenstein

Background: Intrahepatic cholangiocarcinoma (ICC) is a disease with poor prognosis and limited therapeutic options. We investigated the tumor immune microenvironment (TIME) to identify predictors of disease outcome and to explore targets for therapeutic modulation.

Methods: Liver tissue samples were collected during 2008-2019 from patients (n = 139) diagnosed with ICC who underwent curative intent surgery without neoadjuvant chemotherapy. Samples from the discovery cohort (n = 86) were immunohistochemically analyzed on tissue microarrays (TMAs) for the expression of CD68, CD3, CD4, CD8, Foxp3, PD-L1, STAT1, and p-STAT1 in tumor core and stroma areas. Results were digitally analyzed using QuPath software and correlated with clinicopathological characteristics. For validation of TIME-related biomarkers, we performed multiplex imaging mass cytometry (IMC) in a validation cohort (n = 53).

Results: CD68+ cells were the predominant immune cell type in the TIME of ICC. CD4+high T cell density correlated with better overall survival (OS). Prediction modeling together with validation cohort confirmed relevance of CD4+ cells, PD-L1 expression by immune cells in the stroma and N-stage on overall disease outcome. In turn, IMC analyses revealed that silent CD3+CD4+ clusters inversely impacted survival. Among annotated immune cell clusters, PD-L1 was most relevantly expressed by CD4+FoxP3+ cells. A subset of tumors with high density of immune cells ("hot" cluster) correlated with PD-L1 expression and could identify a group of candidates for immune checkpoint inhibition (ICI). Ultimately, higher levels of STAT1 expression were associated with higher lymphocyte infiltration and PD-L1 expression.

Conclusions: These results highlight the importance of CD4+ T cells in immune response against ICC. Secondly, a subset of tumors with "hot" TIME represents potential candidates for ICI, while stimulation of STAT1 pathway could be a potential target to turn "cold" into "hot" TIME in ICC.

背景:肝内胆管癌(ICC肝内胆管癌(ICC)是一种预后不良且治疗方案有限的疾病。我们研究了肿瘤免疫微环境(TIME),以确定疾病预后的预测因素,并探索治疗调节的靶点:2008-2019年期间,我们收集了被诊断为ICC的患者(139人)的肝脏组织样本,这些患者接受了根治性手术,但未接受新辅助化疗。对发现队列中的样本(n = 86)进行组织芯片(TMA)免疫组化分析,检测肿瘤核心区和基质区的 CD68、CD3、CD4、CD8、Foxp3、PD-L1、STAT1 和 p-STAT1 的表达。使用 QuPath 软件对结果进行数字分析,并将其与临床病理特征相关联。为了验证与 TIME 相关的生物标记物,我们在一个验证队列(n = 53)中进行了多重成像质谱(IMC)分析:结果:CD68+细胞是ICC TIME中最主要的免疫细胞类型。CD4+高T细胞密度与较好的总生存期(OS)相关。预测模型和验证队列证实了CD4+细胞、基质中免疫细胞的PD-L1表达和N期对总体疾病预后的相关性。反过来,IMC分析显示,沉默的CD3+CD4+细胞群对生存率有反向影响。在注释的免疫细胞群中,CD4+FoxP3+细胞表达的PD-L1最为相关。免疫细胞密度高的肿瘤亚群("热 "群)与PD-L1的表达相关,可以确定一组免疫检查点抑制剂(ICI)的候选者。最终,较高水平的STAT1表达与较高的淋巴细胞浸润和PD-L1表达相关:这些结果凸显了 CD4+ T 细胞在针对 ICC 的免疫反应中的重要性。其次,具有 "热 "TIME的肿瘤亚群是ICI的潜在候选者,而刺激STAT1通路可能是将ICC中的 "冷 "TIME变为 "热 "TIME的潜在靶点。
{"title":"Clinical prognosticators and targets in the immune microenvironment of intrahepatic cholangiocarcinoma.","authors":"Isis Lozzi, Alexander Arnold, Matthias Barone, Juliette Claire Johnson, Bruno V Sinn, Johannes Eschrich, Pimrapat Gebert, Ruonan Wang, Mengwen Hu, Linda Feldbrügge, Anja Schirmeier, Anja Reutzel-Selke, Thomas Malinka, Felix Krenzien, Wenzel Schöning, Dominik P Modest, Johann Pratschke, Igor M Sauer, Matthäus Felsenstein","doi":"10.1080/2162402X.2024.2406052","DOIUrl":"10.1080/2162402X.2024.2406052","url":null,"abstract":"<p><strong>Background: </strong>Intrahepatic cholangiocarcinoma (ICC) is a disease with poor prognosis and limited therapeutic options. We investigated the tumor immune microenvironment (TIME) to identify predictors of disease outcome and to explore targets for therapeutic modulation.</p><p><strong>Methods: </strong>Liver tissue samples were collected during 2008-2019 from patients (<i>n</i> = 139) diagnosed with ICC who underwent curative intent surgery without neoadjuvant chemotherapy. Samples from the discovery cohort (<i>n</i> = 86) were immunohistochemically analyzed on tissue microarrays (TMAs) for the expression of CD68, CD3, CD4, CD8, Foxp3, PD-L1, STAT1, and p-STAT1 in tumor core and stroma areas. Results were digitally analyzed using QuPath software and correlated with clinicopathological characteristics. For validation of TIME-related biomarkers, we performed multiplex imaging mass cytometry (IMC) in a validation cohort (<i>n</i> = 53).</p><p><strong>Results: </strong>CD68+ cells were the predominant immune cell type in the TIME of ICC. CD4+<sup>high</sup> T cell density correlated with better overall survival (OS). Prediction modeling together with validation cohort confirmed relevance of CD4+ cells, PD-L1 expression by immune cells in the stroma and N-stage on overall disease outcome. In turn, IMC analyses revealed that silent CD3+CD4+ clusters inversely impacted survival. Among annotated immune cell clusters, PD-L1 was most relevantly expressed by CD4+FoxP3+ cells. A subset of tumors with high density of immune cells (\"hot\" cluster) correlated with PD-L1 expression and could identify a group of candidates for immune checkpoint inhibition (ICI). Ultimately, higher levels of STAT1 expression were associated with higher lymphocyte infiltration and PD-L1 expression.</p><p><strong>Conclusions: </strong>These results highlight the importance of CD4+ T cells in immune response against ICC. Secondly, a subset of tumors with \"hot\" TIME represents potential candidates for ICI, while stimulation of STAT1 pathway could be a potential target to turn \"cold\" into \"hot\" TIME in ICC.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11445892/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142367086","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Enhancing T-cell recruitment in renal cell carcinoma with cytokine-armed adenoviruses. 用细胞因子武装的腺病毒增强肾细胞癌中的 T 细胞募集。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-25 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2407532
Michaela Feodoroff, Firas Hamdan, Gabriella Antignani, Sara Feola, Manlio Fusciello, Salvatore Russo, Jacopo Chiaro, Katja Välimäki, Teijo Pellinen, Rui M Branca, Janne Lehtiö, Federica D Alessio, Paolo Bottega, Virpi Stigzelius, Janita Sandberg, Jonna Clancy, Jukka Partanen, Minna Malmstedt, Antti Rannikko, Vilja Pietiäinen, Mikaela Grönholm, Vincenzo Cerullo

Immunotherapy has emerged as a promising approach for cancer treatment, with oncolytic adenoviruses showing power as immunotherapeutic agents. In this study, we investigated the immunotherapeutic potential of an adenovirus construct expressing CXCL9, CXCL10, or IL-15 in clear cell renal cell carcinoma (ccRCC) tumor models. Our results demonstrated robust cytokine secretion upon viral treatment, suggesting effective transgene expression. Subsequent analysis using resistance-based transwell migration and microfluidic chip assays demonstrated increased T-cell migration in response to chemokine secretion by infected cells in both 2D and 3D cell models. Flow cytometry analysis revealed CXCR3 receptor expression across T-cell subsets, with the highest percentage found on CD8+ T-cells, underscoring their key role in immune cell migration. Alongside T-cells, we also detected NK-cells in the tumors of immunocompromised mice treated with cytokine-encoding adenoviruses. Furthermore, we identified potential immunogenic antigens that may enhance the efficacy and specificity of our armed oncolytic adenoviruses in ccRCC. Overall, our findings using ccRCC cell line, in vivo humanized mice, physiologically relevant PDCs in 2D and patient-derived organoids (PDOs) in 3D suggest that chemokine-armed adenoviruses hold promise for enhancing T-cell migration and improving immunotherapy outcomes in ccRCC. Our study contributes to the development of more effective ccRCC treatment strategies by elucidating immune cell infiltration and activation mechanisms within the tumor microenvironment (TME) and highlights the usefulness of PDOs for predicting clinical relevance and validating novel immunotherapeutic approaches. Overall, our research offers insights into the rational design and optimization of viral-based immunotherapies for ccRCC.

免疫疗法已成为一种前景广阔的癌症治疗方法,溶瘤腺病毒作为免疫治疗药物显示出强大的威力。在本研究中,我们研究了表达 CXCL9、CXCL10 或 IL-15 的腺病毒构建体在透明细胞肾细胞癌(ccRCC)肿瘤模型中的免疫治疗潜力。我们的结果表明,病毒处理后细胞因子分泌旺盛,表明转基因表达有效。随后使用基于抗性的透孔迁移和微流控芯片测定法进行的分析表明,在二维和三维细胞模型中,T细胞迁移增加是对感染细胞分泌趋化因子的反应。流式细胞术分析表明,CXCR3受体在各T细胞亚群中均有表达,其中CD8+ T细胞的表达量最高,这表明它们在免疫细胞迁移中发挥着关键作用。除了 T 细胞,我们还在接受细胞因子编码腺病毒治疗的免疫缺陷小鼠肿瘤中检测到了 NK 细胞。此外,我们还发现了潜在的免疫原性抗原,这些抗原可能会提高我们的武装溶瘤腺病毒在ccRCC中的疗效和特异性。总之,我们使用ccRCC细胞系、体内人源化小鼠、二维生理学相关PDCs和三维患者衍生器官组织(PDOs)的研究结果表明,趋化因子武装腺病毒有望增强T细胞迁移,改善ccRCC的免疫治疗效果。我们的研究通过阐明肿瘤微环境(TME)中的免疫细胞浸润和激活机制,为开发更有效的ccRCC治疗策略做出了贡献,并强调了PDOs在预测临床相关性和验证新型免疫治疗方法方面的有用性。总之,我们的研究为合理设计和优化基于病毒的 ccRCC 免疫疗法提供了启示。
{"title":"Enhancing T-cell recruitment in renal cell carcinoma with cytokine-armed adenoviruses.","authors":"Michaela Feodoroff, Firas Hamdan, Gabriella Antignani, Sara Feola, Manlio Fusciello, Salvatore Russo, Jacopo Chiaro, Katja Välimäki, Teijo Pellinen, Rui M Branca, Janne Lehtiö, Federica D Alessio, Paolo Bottega, Virpi Stigzelius, Janita Sandberg, Jonna Clancy, Jukka Partanen, Minna Malmstedt, Antti Rannikko, Vilja Pietiäinen, Mikaela Grönholm, Vincenzo Cerullo","doi":"10.1080/2162402X.2024.2407532","DOIUrl":"10.1080/2162402X.2024.2407532","url":null,"abstract":"<p><p>Immunotherapy has emerged as a promising approach for cancer treatment, with oncolytic adenoviruses showing power as immunotherapeutic agents. In this study, we investigated the immunotherapeutic potential of an adenovirus construct expressing CXCL9, CXCL10, or IL-15 in clear cell renal cell carcinoma (ccRCC) tumor models. Our results demonstrated robust cytokine secretion upon viral treatment, suggesting effective transgene expression. Subsequent analysis using resistance-based transwell migration and microfluidic chip assays demonstrated increased T-cell migration in response to chemokine secretion by infected cells in both 2D and 3D cell models. Flow cytometry analysis revealed CXCR3 receptor expression across T-cell subsets, with the highest percentage found on CD8+ T-cells, underscoring their key role in immune cell migration. Alongside T-cells, we also detected NK-cells in the tumors of immunocompromised mice treated with cytokine-encoding adenoviruses. Furthermore, we identified potential immunogenic antigens that may enhance the efficacy and specificity of our armed oncolytic adenoviruses in ccRCC. Overall, our findings using ccRCC cell line, <i>in vivo</i> humanized mice, physiologically relevant PDCs in 2D and patient-derived organoids (PDOs) in 3D suggest that chemokine-armed adenoviruses hold promise for enhancing T-cell migration and improving immunotherapy outcomes in ccRCC. Our study contributes to the development of more effective ccRCC treatment strategies by elucidating immune cell infiltration and activation mechanisms within the tumor microenvironment (TME) and highlights the usefulness of PDOs for predicting clinical relevance and validating novel immunotherapeutic approaches. Overall, our research offers insights into the rational design and optimization of viral-based immunotherapies for ccRCC.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11441019/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142356459","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Immune priming and induction of tertiary lymphoid structures in a cord-blood humanized mouse model of gastrointestinal stromal tumor. 脐带血人源化胃肠道间质瘤小鼠模型中的免疫启动和三级淋巴结构诱导。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-09-22 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2406576
Bo He, Larissa Dymond, Kira H Wood, Edward R Bastow, Jiulia Satiaputra, Ji Li, Anna Johansson-Percival, Juliana Hamzah, M Priyanthi Kumarasinghe, Mohammed Ballal, Jonathan Foo, Mikael Johansson, Hooi C Ee, Scott W White, Louise Winteringham, Ruth Ganss

Gastrointestinal stromal tumors (GISTs) harbor diverse immune cell populations but so far immunotherapy in patients has been disappointing. Here, we established cord blood humanized mouse models of localized and disseminated GIST to explore the remodeling of the tumor environment for improved immunotherapy. Specifically, we assessed the ability of a cancer vascular targeting peptide (VTP) to bind to mouse and patient GIST angiogenic blood vessels and deliver the TNF superfamily member LIGHT (TNFS14) into tumors. LIGHT-VTP treatment of GIST in humanized mice improved vascular function and tumor oxygenation, which correlated with an overall increase in intratumoral human effector T cells. Concomitant with LIGHT-mediated vascular remodeling, we observed intratumoral high endothelial venules (HEVs) and tertiary lymphoid structures (TLS), which resemble spontaneous TLS found in GIST patients. Thus, by overcoming the limitations of immunodeficient xenograft models, we demonstrate the therapeutic feasibility of vascular targeting and immune priming in human GIST. Since TLS positively correlate with patient prognosis and improved response to immune checkpoint inhibition, vascular LIGHT targeting in GIST is a highly translatable approach to improve immunotherapeutic outcomes.

胃肠道间质瘤(GIST)蕴藏着多种多样的免疫细胞群,但迄今为止对患者的免疫治疗效果令人失望。在这里,我们建立了脐带血人源化小鼠局部和播散 GIST 模型,探索重塑肿瘤环境以改善免疫疗法。具体来说,我们评估了癌症血管靶向肽(VTP)与小鼠和患者GIST血管生成血管结合并将TNF超家族成员LIGHT(TNFS14)递送到肿瘤的能力。LIGHT-VTP可改善人源化小鼠的血管功能和肿瘤氧合,这与瘤内人类效应T细胞的整体增加有关。在LIGHT介导的血管重塑的同时,我们观察到了瘤内高内皮静脉(HEVs)和三级淋巴结构(TLS),这与GIST患者中发现的自发性TLS相似。因此,通过克服免疫缺陷异种移植模型的局限性,我们证明了血管靶向和免疫激活在人类 GIST 中的治疗可行性。由于TLS与患者的预后和对免疫检查点抑制的反应改善呈正相关,因此GIST中的血管LIGHT靶向是一种可转化性很高的改善免疫治疗效果的方法。
{"title":"Immune priming and induction of tertiary lymphoid structures in a cord-blood humanized mouse model of gastrointestinal stromal tumor.","authors":"Bo He, Larissa Dymond, Kira H Wood, Edward R Bastow, Jiulia Satiaputra, Ji Li, Anna Johansson-Percival, Juliana Hamzah, M Priyanthi Kumarasinghe, Mohammed Ballal, Jonathan Foo, Mikael Johansson, Hooi C Ee, Scott W White, Louise Winteringham, Ruth Ganss","doi":"10.1080/2162402X.2024.2406576","DOIUrl":"10.1080/2162402X.2024.2406576","url":null,"abstract":"<p><p>Gastrointestinal stromal tumors (GISTs) harbor diverse immune cell populations but so far immunotherapy in patients has been disappointing. Here, we established cord blood humanized mouse models of localized and disseminated GIST to explore the remodeling of the tumor environment for improved immunotherapy. Specifically, we assessed the ability of a cancer vascular targeting peptide (VTP) to bind to mouse and patient GIST angiogenic blood vessels and deliver the TNF superfamily member LIGHT (TNFS14) into tumors. LIGHT-VTP treatment of GIST in humanized mice improved vascular function and tumor oxygenation, which correlated with an overall increase in intratumoral human effector T cells. Concomitant with LIGHT-mediated vascular remodeling, we observed intratumoral high endothelial venules (HEVs) and tertiary lymphoid structures (TLS), which resemble spontaneous TLS found in GIST patients. Thus, by overcoming the limitations of immunodeficient xenograft models, we demonstrate the therapeutic feasibility of vascular targeting and immune priming in human GIST. Since TLS positively correlate with patient prognosis and improved response to immune checkpoint inhibition, vascular LIGHT targeting in GIST is a highly translatable approach to improve immunotherapeutic outcomes.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5,"publicationDate":"2024-09-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11418220/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142308843","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
CD4+ tumor-infiltrating lymphocytes secreting T cell-engagers induce regression of autologous patient-derived non-small cell lung cancer xenografts. 分泌 T 细胞诱导因子的 CD4+ 肿瘤浸润淋巴细胞可诱导自体非小细胞肺癌异种移植物消退。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-27 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2392897
Anaïs Jiménez-Reinoso, Magdalena Molero-Abraham, Cristina Cirauqui, Belén Blanco, Eva M Garrido-Martin, Daniel Nehme-Álvarez, Carmen Domínguez-Alonso, Ángel Ramírez-Fernández, Laura Díez-Alonso, Ángel Nuñez-Buiza, África González-Murillo, Raquel Tobes, Eduardo Pareja, Manuel Ramírez-Orellana, José Luis Rodriguez-Peralto, Irene Ferrer, Jon Zugazagoitia, Luis Paz-Ares, Luis Álvarez-Vallina

Adoptive transfer of tumor-infiltrating lymphocytes (TIL) has shown remarkable results in melanoma, but only modest clinical benefits in other cancers, even after TIL have been genetically modified to improve their tumor homing, cytotoxic potential or overcome cell exhaustion. The required ex vivo TIL expansion process may induce changes in the T cell clonal composition, which could likely compromise the tumor reactivity of TIL preparations and ultimately the success of TIL therapy. A promising approach based on the production of bispecific T cell-engagers (TCE) by engineered T cells (STAb-T therapy) improves the efficacy of current T cell redirection strategies against tumor-associated antigens in hematological tumors. We studied the TCRβ repertoire in non-small cell lung cancer (NSCLC) tumors and in ex vivo expanded TIL from two unrelated patients. We generated TIL secreting anti-epidermal growth factor receptor (EGFR) × anti-CD3 TCE (TILSTAb) and tested their antitumor efficacy in vitro and in vivo using a NSCLC patient-derived xenograft (PDX) model in which tumor fragments and TIL from the same patient were transplanted into hIL-2 NOG mice. We confirmed that the standard TIL expansion protocol promotes the loss of tumor-dominant T cell clones and the overgrowth of virus-reactive TCR clonotypes that were marginally detectable in primary tumors. We demonstrated the antitumor activity of TILSTAb both in vitro and in vivo when administered intratumorally and systemically in an autologous immune-humanized PDX EGFR+ NSCLC mouse model, where tumor regression was mediated by TCE-redirected CD4+ TIL bearing non-tumor dominant clonotypes.

肿瘤浸润淋巴细胞(TIL)的采用性转移在黑色素瘤中取得了显著效果,但在其他癌症中的临床疗效却微乎其微,即使在对 TIL 进行基因修饰以改善其肿瘤归巢性、细胞毒性潜力或克服细胞衰竭之后也是如此。所需的体内外 TIL 扩增过程可能会引起 T 细胞克隆组成的变化,这可能会影响 TIL 制剂的肿瘤反应性,最终影响 TIL 治疗的成功。一种很有前景的方法是通过工程T细胞产生双特异性T细胞激活剂(TCE)(STAb-T疗法),这种方法提高了目前针对血液肿瘤中肿瘤相关抗原的T细胞重定向策略的疗效。我们研究了非小细胞肺癌(NSCLC)肿瘤中的 TCRβ 重排,以及来自两名非亲缘关系患者的体外扩增 TIL。我们生成了分泌抗表皮生长因子受体(EGFR)×抗CD3 TCE(TILSTAb)的TIL,并使用NSCLC患者异种移植(PDX)模型测试了它们在体外和体内的抗肿瘤疗效,该模型是将来自同一患者的肿瘤片段和TIL移植到hIL-2 NOG小鼠体内。我们证实,标准的 TIL 扩增方案会导致肿瘤主导型 T 细胞克隆的丧失和病毒反应型 TCR 克隆型的过度生长,而这些克隆型在原发性肿瘤中几乎检测不到。在自体免疫人源化 PDX EGFR+ NSCLC 小鼠模型中,我们证实了 TILSTAb 在瘤内和全身给药的体外和体内抗肿瘤活性。
{"title":"CD4<sup>+</sup> tumor-infiltrating lymphocytes secreting T cell-engagers induce regression of autologous patient-derived non-small cell lung cancer xenografts.","authors":"Anaïs Jiménez-Reinoso, Magdalena Molero-Abraham, Cristina Cirauqui, Belén Blanco, Eva M Garrido-Martin, Daniel Nehme-Álvarez, Carmen Domínguez-Alonso, Ángel Ramírez-Fernández, Laura Díez-Alonso, Ángel Nuñez-Buiza, África González-Murillo, Raquel Tobes, Eduardo Pareja, Manuel Ramírez-Orellana, José Luis Rodriguez-Peralto, Irene Ferrer, Jon Zugazagoitia, Luis Paz-Ares, Luis Álvarez-Vallina","doi":"10.1080/2162402X.2024.2392897","DOIUrl":"10.1080/2162402X.2024.2392897","url":null,"abstract":"<p><p>Adoptive transfer of tumor-infiltrating lymphocytes (TIL) has shown remarkable results in melanoma, but only modest clinical benefits in other cancers, even after TIL have been genetically modified to improve their tumor homing, cytotoxic potential or overcome cell exhaustion. The required <i>ex vivo</i> TIL expansion process may induce changes in the T cell clonal composition, which could likely compromise the tumor reactivity of TIL preparations and ultimately the success of TIL therapy. A promising approach based on the production of bispecific T cell-engagers (TCE) by engineered T cells (STAb-T therapy) improves the efficacy of current T cell redirection strategies against tumor-associated antigens in hematological tumors. We studied the TCRβ repertoire in non-small cell lung cancer (NSCLC) tumors and in <i>ex vivo</i> expanded TIL from two unrelated patients. We generated TIL secreting anti-epidermal growth factor receptor (EGFR) × anti-CD3 TCE (TIL<sup>STAb</sup>) and tested their antitumor efficacy <i>in vitro</i> and <i>in vivo</i> using a NSCLC patient-derived xenograft (PDX) model in which tumor fragments and TIL from the same patient were transplanted into <i>hIL-2</i> NOG mice. We confirmed that the standard TIL expansion protocol promotes the loss of tumor-dominant T cell clones and the overgrowth of virus-reactive TCR clonotypes that were marginally detectable in primary tumors. We demonstrated the antitumor activity of TIL<sup>STAb</sup> both <i>in vitro</i> and <i>in vivo</i> when administered intratumorally and systemically in an autologous immune-humanized PDX EGFR<sup>+</sup> NSCLC mouse model, where tumor regression was mediated by TCE-redirected CD4<sup>+</sup> TIL bearing non-tumor dominant clonotypes.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5,"publicationDate":"2024-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11352715/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142113732","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Overexpression of CCL-20 and CXCL-8 genes enhances tumor escape and resistance to cemiplimab, a programmed cell death protein-1 (PD-1) inhibitor, in patients with locally advanced and metastatic cutaneous squamous cell carcinoma. CCL-20和CXCL-8基因的过度表达增强了局部晚期和转移性皮肤鳞状细胞癌患者的肿瘤逃逸能力和对程序性细胞死亡蛋白-1(PD-1)抑制剂cemiplimab的耐药性。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-26 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2388315
Vincenzo De Falco, Stefania Napolitano, Renato Franco, Federica Zito Marino, Luigi Formisano, Daniela Esposito, Gabriella Suarato, Rossella Napolitano, Alfonso Esposito, Francesco Caraglia, Maria Cristina Giugliano, Eleonora Cioli, Vincenzo Famiglietti, Roberto Bianco, Giuseppe Argenziano, Andrea Ronchi, Davide Ciardiello, Valerio Nardone, Emma D'Ippolito, Sara Del Tufo, Fortunato Ciardiello, Teresa Troiani

Cemiplimab has demonstrated relevant clinical activity in cutaneous squamous cell carcinoma (cSCC) but mechanisms of primary and acquired resistance to immunotherapy are still unknown. We collected clinical data from locally advanced and/or metastatic cSSC patients treated with cemiplimab in two Italian University centers. In addition, gene expression analysis by using Nanostring Technologies platform to evaluate 770 cancer- and immune-related genes on 20 tumor tissue samples (9 responders and 11 non-responders to cemiplimab) was performed. We enrolled 81 patients with a median age of 82 years. After 16.4 months of median follow-up, 12- and 24-months PFS were 53% and 42%, respectively; while 12- and 24-months OS were 71% and 61%, respectively. Treatment was well tolerated. Overall response rate (ORR) was 58%, with a disease control rate (DCR) of 77.8%. The difference between genes expressed in responder versus non-responder patient samples was substantial, particularly for genes involved in immune system regulation. Cemiplimab-resistant tumors were associated with over-expression of CCL-20 and CXCL-8. Cemiplimab confirmed efficacy and safety data in real-life cSCC patients. Overexpression of CCL-20 and CXCL-8 could represent biomarkers of lack of response to immunotherapy.

塞米普利姆单抗已在皮肤鳞状细胞癌(cSCC)中显示出相关的临床活性,但免疫疗法的原发性和获得性抗药性机制仍不清楚。我们收集了意大利两所大学中心接受塞米普利单抗治疗的局部晚期和/或转移性 cSSC 患者的临床数据。此外,我们还使用 Nanostring Technologies 平台进行了基因表达分析,评估了 20 份肿瘤组织样本(9 份对赛美普利单抗有反应,11 份无反应)中的 770 个癌症和免疫相关基因。我们共招募了 81 名患者,中位年龄为 82 岁。经过16.4个月的中位随访,12个月和24个月的PFS分别为53%和42%;12个月和24个月的OS分别为71%和61%。治疗耐受性良好。总体反应率(ORR)为58%,疾病控制率(DCR)为77.8%。应答患者与非应答患者样本中表达的基因差异很大,尤其是涉及免疫系统调节的基因。对塞米单抗耐药的肿瘤与 CCL-20 和 CXCL-8 的过度表达有关。塞米普利姆单抗证实了在现实生活中的 cSCC 患者中的疗效和安全性数据。CCL-20和CXCL-8的过度表达可能是对免疫疗法缺乏反应的生物标志物。
{"title":"Overexpression of CCL-20 and CXCL-8 genes enhances tumor escape and resistance to cemiplimab, a programmed cell death protein-1 (PD-1) inhibitor, in patients with locally advanced and metastatic cutaneous squamous cell carcinoma.","authors":"Vincenzo De Falco, Stefania Napolitano, Renato Franco, Federica Zito Marino, Luigi Formisano, Daniela Esposito, Gabriella Suarato, Rossella Napolitano, Alfonso Esposito, Francesco Caraglia, Maria Cristina Giugliano, Eleonora Cioli, Vincenzo Famiglietti, Roberto Bianco, Giuseppe Argenziano, Andrea Ronchi, Davide Ciardiello, Valerio Nardone, Emma D'Ippolito, Sara Del Tufo, Fortunato Ciardiello, Teresa Troiani","doi":"10.1080/2162402X.2024.2388315","DOIUrl":"10.1080/2162402X.2024.2388315","url":null,"abstract":"<p><p>Cemiplimab has demonstrated relevant clinical activity in cutaneous squamous cell carcinoma (cSCC) but mechanisms of primary and acquired resistance to immunotherapy are still unknown. We collected clinical data from locally advanced and/or metastatic cSSC patients treated with cemiplimab in two Italian University centers. In addition, gene expression analysis by using Nanostring Technologies platform to evaluate 770 cancer- and immune-related genes on 20 tumor tissue samples (9 responders and 11 non-responders to cemiplimab) was performed. We enrolled 81 patients with a median age of 82 years. After 16.4 months of median follow-up, 12- and 24-months PFS were 53% and 42%, respectively; while 12- and 24-months OS were 71% and 61%, respectively. Treatment was well tolerated. Overall response rate (ORR) was 58%, with a disease control rate (DCR) of 77.8%. The difference between genes expressed in responder versus non-responder patient samples was substantial, particularly for genes involved in immune system regulation. Cemiplimab-resistant tumors were associated with over-expression of CCL-20 and CXCL-8. Cemiplimab confirmed efficacy and safety data in real-life cSCC patients. Overexpression of CCL-20 and CXCL-8 could represent biomarkers of lack of response to immunotherapy.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5,"publicationDate":"2024-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11352706/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142113734","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Metabolic regulation of the mitochondrial immune checkpoint. 线粒体免疫检查点的代谢调节。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-26 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2394247
David C Montrose, Suchandrima Saha, Lorenzo Galluzzi

Disrupting mitochondrial function in malignant cells is a promising strategy to enhance anticancer immunity. We have recently demonstrated that depriving colorectal cancer cells of serine results in mitochondrial dysfunction coupled with the cytosolic accumulation of mitochondrial DNA and consequent activation of CGAS- and STING-dependent tumor-targeting immune responses.

破坏恶性细胞的线粒体功能是一种很有前景的增强抗癌免疫力的策略。我们最近证明,剥夺结直肠癌细胞中的丝氨酸会导致线粒体功能障碍以及线粒体 DNA 的胞浆积累,从而激活依赖 CGAS 和 STING 的肿瘤靶向免疫反应。
{"title":"Metabolic regulation of the mitochondrial immune checkpoint.","authors":"David C Montrose, Suchandrima Saha, Lorenzo Galluzzi","doi":"10.1080/2162402X.2024.2394247","DOIUrl":"10.1080/2162402X.2024.2394247","url":null,"abstract":"<p><p>Disrupting mitochondrial function in malignant cells is a promising strategy to enhance anticancer immunity. We have recently demonstrated that depriving colorectal cancer cells of serine results in mitochondrial dysfunction coupled with the cytosolic accumulation of mitochondrial DNA and consequent activation of CGAS- and STING-dependent tumor-targeting immune responses.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5,"publicationDate":"2024-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11352702/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142113733","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Local therapy with combination TLR agonists stimulates systemic anti-tumor immunity and sensitizes tumors to immune checkpoint blockade. 联合 TLR 激动剂的局部疗法可刺激全身抗肿瘤免疫,并使肿瘤对免疫检查点阻断剂敏感。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-22 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2395067
Francois Xavier Rwandamuriye, Tao Wang, Hanfu Zhang, Omar Elaskalani, Jorren Kuster, Xueting Ye, Breana Vitali, Juliët Schreurs, M Lizeth Orozco Morales, Marck Norret, Cameron W Evans, Rachael M Zemek, K Swaminathan Iyer, W Joost Lesterhuis, Ben Wylie

Toll-like receptor (TLR) agonists are being developed as anti-cancer therapeutics due to their potent immunostimulatory properties. However, clinical trials testing TLR agonists as monotherapy have often failed to demonstrate significant improvement over standard of care. We hypothesized that the anti-cancer efficacy of TLR agonist immunotherapy could be improved by combinatorial approaches. To prevent increased toxicity, often seen with systemic combination therapies, we developed a hydrogel to deliver TLR agonist combinations at low doses, locally, during cancer debulking surgery. Using tumor models of WEHI 164 and bilateral M3-9-M sarcoma and CT26 colon carcinoma, we assessed the efficacy of pairwise combinations of poly(I:C), R848, and CpG in controlling local and distant tumor growth. We show that combination of the TLR3 agonist poly(I:C) and TLR7/8 agonist R848 drives anti-tumor immunity against local and distant tumors. In addition, combination of local poly(I:C) and R848 sensitized tumors to systemic immune checkpoint blockade, improving tumor control. Mechanistically, we demonstrate that local therapy with poly(I:C) and R848 recruits inflammatory monocytes to the tumor draining lymph nodes early in the anti-tumor response. Finally, we provide proof of concept for intraoperative delivery of poly(I:C) and R848 together via a surgically applicable biodegradable hydrogel.

Toll样受体(TLR)激动剂因其强大的免疫刺激特性,正被开发为抗癌疗法。然而,将 TLR 激动剂作为单一疗法进行测试的临床试验往往无法证明其疗效明显优于标准疗法。我们假设,TLR 激动剂免疫疗法的抗癌效果可以通过组合方法得到改善。为了防止全身性联合疗法经常出现的毒性增加,我们开发了一种水凝胶,用于在癌症剥离手术中以低剂量局部递送 TLR 激动剂组合。利用 WEHI 164 和双侧 M3-9-M 肉瘤以及 CT26 结肠癌的肿瘤模型,我们评估了 poly(I:C)、R848 和 CpG 配对组合在控制局部和远处肿瘤生长方面的疗效。我们的研究表明,TLR3 激动剂 poly(I:C) 和 TLR7/8 激动剂 R848 的组合能增强对局部和远处肿瘤的抗肿瘤免疫力。此外,局部聚(I:C)和 R848 的联合使用使肿瘤对全身免疫检查点阻断剂敏感,从而改善了肿瘤控制。从机理上讲,我们证明了聚(I:C)和R848的局部疗法能在抗肿瘤反应早期将炎性单核细胞招募到肿瘤引流淋巴结。最后,我们提供了通过手术适用的生物可降解水凝胶术中递送聚(I:C)和 R848 的概念验证。
{"title":"Local therapy with combination TLR agonists stimulates systemic anti-tumor immunity and sensitizes tumors to immune checkpoint blockade.","authors":"Francois Xavier Rwandamuriye, Tao Wang, Hanfu Zhang, Omar Elaskalani, Jorren Kuster, Xueting Ye, Breana Vitali, Juliët Schreurs, M Lizeth Orozco Morales, Marck Norret, Cameron W Evans, Rachael M Zemek, K Swaminathan Iyer, W Joost Lesterhuis, Ben Wylie","doi":"10.1080/2162402X.2024.2395067","DOIUrl":"10.1080/2162402X.2024.2395067","url":null,"abstract":"<p><p>Toll-like receptor (TLR) agonists are being developed as anti-cancer therapeutics due to their potent immunostimulatory properties. However, clinical trials testing TLR agonists as monotherapy have often failed to demonstrate significant improvement over standard of care. We hypothesized that the anti-cancer efficacy of TLR agonist immunotherapy could be improved by combinatorial approaches. To prevent increased toxicity, often seen with systemic combination therapies, we developed a hydrogel to deliver TLR agonist combinations at low doses, locally, during cancer debulking surgery. Using tumor models of WEHI 164 and bilateral M3-9-M sarcoma and CT26 colon carcinoma, we assessed the efficacy of pairwise combinations of poly(I:C), R848, and CpG in controlling local and distant tumor growth. We show that combination of the TLR3 agonist poly(I:C) and TLR7/8 agonist R848 drives anti-tumor immunity against local and distant tumors. In addition, combination of local poly(I:C) and R848 sensitized tumors to systemic immune checkpoint blockade, improving tumor control. Mechanistically, we demonstrate that local therapy with poly(I:C) and R848 recruits inflammatory monocytes to the tumor draining lymph nodes early in the anti-tumor response. Finally, we provide proof of concept for intraoperative delivery of poly(I:C) and R848 together via a surgically applicable biodegradable hydrogel.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11346538/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142074255","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Tertiary lymphoid structure-related immune infiltrates in NSCLC tumor lesions correlate with low tumor-reactivity of TIL products. NSCLC 肿瘤病灶中与三级淋巴结构相关的免疫浸润与 TIL 产物的低肿瘤反应性相关。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-22 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2392898
Suzanne M Castenmiller, Nandhini Kanagasabesan, Aurélie Guislain, Benoît P Nicolet, Marleen M van Loenen, Kim Monkhorst, Alexander A F A Veenhof, Egbert F Smit, Koen J Hartemink, John B A G Haanen, Rosa de Groot, Monika C Wolkers

Adoptive transfer of tumor infiltrating lymphocytes (TIL therapy) has proven highly effective for treating solid cancers, including non-small cell lung cancer (NSCLC). However, not all patients benefit from this therapy for yet unknown reasons. Defining markers that correlate with high tumor-reactivity of the autologous TIL products is thus key for achieving better tailored immunotherapies. We questioned whether the composition of immune cell infiltrates correlated with the tumor-reactivity of expanded TIL products. Unbiased flow cytometry analysis of immune cell infiltrates of 26 early-stage and 20 late-stage NSCLC tumor lesions was used for correlations with the T cell differentiation and activation status, and with the expansion rate and anti-tumor response of generated TIL products. The composition of tumor immune infiltrates was highly variable between patients. Spearman's Rank Correlation revealed that high B cell infiltration negatively correlated with the tumor-reactivity of the patient's expanded TIL products, as defined by cytokine production upon exposure to autologous tumor digest. In-depth analysis revealed that tumor lesions with high B cell infiltrates contained tertiary lymphoid structure (TLS)-related immune infiltrates, including BCL6+ antibody-secreting B cells, IgD+BCL6+ B cells and CXCR5+BLC6+ CD4+ T cells, and higher percentages of naïve CD8+ T cells. In conclusion, the composition of immune cell infiltrates in NSCLC tumors associates with the functionality of the expanded TIL product. Our findings may thus help improve patient selection for TIL therapy.

事实证明,肿瘤浸润淋巴细胞的采纳转移(TIL疗法)对治疗包括非小细胞肺癌(NSCLC)在内的实体瘤非常有效。然而,并非所有患者都能从这种疗法中获益,原因尚不清楚。因此,确定与自体 TIL 产物的高肿瘤反应性相关的标记物是实现更好的定制免疫疗法的关键。我们对免疫细胞浸润的组成是否与扩增 TIL 产物的肿瘤反应性相关提出了疑问。我们对 26 例早期和 20 例晚期 NSCLC 肿瘤病灶的免疫细胞浸润进行了无偏流式细胞术分析,以确定其与 T 细胞分化和活化状态、扩增率以及生成的 TIL 产物的抗肿瘤反应之间的相关性。不同患者的肿瘤免疫浸润组成差异很大。斯皮尔曼等级相关性显示,高B细胞浸润与患者扩增的TIL产物的肿瘤反应性呈负相关,其定义是暴露于自体肿瘤消化液时产生的细胞因子。深入分析发现,B细胞浸润较高的肿瘤病灶含有三级淋巴结构(TLS)相关的免疫浸润,包括分泌抗体的BCL6+ B细胞、IgD+BCL6+ B细胞和CXCR5+BLC6+ CD4+ T细胞,以及较高比例的幼稚CD8+ T细胞。总之,NSCLC 肿瘤中免疫细胞浸润的组成与扩增的 TIL 产物的功能有关。因此,我们的发现可能有助于改善TIL疗法的患者选择。
{"title":"Tertiary lymphoid structure-related immune infiltrates in NSCLC tumor lesions correlate with low tumor-reactivity of TIL products.","authors":"Suzanne M Castenmiller, Nandhini Kanagasabesan, Aurélie Guislain, Benoît P Nicolet, Marleen M van Loenen, Kim Monkhorst, Alexander A F A Veenhof, Egbert F Smit, Koen J Hartemink, John B A G Haanen, Rosa de Groot, Monika C Wolkers","doi":"10.1080/2162402X.2024.2392898","DOIUrl":"10.1080/2162402X.2024.2392898","url":null,"abstract":"<p><p>Adoptive transfer of tumor infiltrating lymphocytes (TIL therapy) has proven highly effective for treating solid cancers, including non-small cell lung cancer (NSCLC). However, not all patients benefit from this therapy for yet unknown reasons. Defining markers that correlate with high tumor-reactivity of the autologous TIL products is thus key for achieving better tailored immunotherapies. We questioned whether the composition of immune cell infiltrates correlated with the tumor-reactivity of expanded TIL products. Unbiased flow cytometry analysis of immune cell infiltrates of 26 early-stage and 20 late-stage NSCLC tumor lesions was used for correlations with the T cell differentiation and activation status, and with the expansion rate and anti-tumor response of generated TIL products. The composition of tumor immune infiltrates was highly variable between patients. Spearman's Rank Correlation revealed that high B cell infiltration negatively correlated with the tumor-reactivity of the patient's expanded TIL products, as defined by cytokine production upon exposure to autologous tumor digest. In-depth analysis revealed that tumor lesions with high B cell infiltrates contained tertiary lymphoid structure (TLS)-related immune infiltrates, including BCL6<sup>+</sup> antibody-secreting B cells, IgD<sup>+</sup>BCL6<sup>+</sup> B cells and CXCR5<sup>+</sup>BLC6<sup>+</sup> CD4<sup>+</sup> T cells, and higher percentages of naïve CD8<sup>+</sup> T cells. In conclusion, the composition of immune cell infiltrates in NSCLC tumors associates with the functionality of the expanded TIL product. Our findings may thus help improve patient selection for TIL therapy.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11346574/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142074256","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Secreted factors from M1 macrophages drive prostate cancer stem cell plasticity by upregulating NANOG, SOX2, and CD44 through NFκB-signaling. M1巨噬细胞分泌的因子通过NFκB信号上调NANOG、SOX2和CD44,驱动前列腺癌干细胞的可塑性。
IF 6.5 2区 医学 Q1 IMMUNOLOGY Pub Date : 2024-08-21 eCollection Date: 2024-01-01 DOI: 10.1080/2162402X.2024.2393442
Kirsi Kainulainen, Einari A Niskanen, Johanna Kinnunen, Kaisa Mäki-Mantila, Kiia Hartikainen, Ville Paakinaho, Marjo Malinen, Kirsi Ketola, Sanna Pasonen-Seppänen

The inflammatory tumor microenvironment (TME) is a key driver for tumor-promoting processes. Tumor-associated macrophages are one of the main immune cell types in the TME and their increased density is related to poor prognosis in prostate cancer. Here, we investigated the influence of pro-inflammatory (M1) and immunosuppressive (M2) macrophages on prostate cancer lineage plasticity. Our findings reveal that M1 macrophage secreted factors upregulate genes related to stemness while downregulating genes associated with androgen response in prostate cancer cells. The expression of cancer stem cell (CSC) plasticity markers NANOG, KLF4, SOX2, OCT4, and CD44 was stimulated by the secreted factors from M1 macrophages. Moreover, AR and its target gene PSA were observed to be suppressed in LNCaP cells treated with secreted factors from M1 macrophages. Inhibition of NFκB signaling using the IKK16 inhibitor resulted in downregulation of NANOG, SOX2, and CD44 and CSC plasticity. Our study highlights that the secreted factors from M1 macrophages drive prostate cancer cell plasticity by upregulating the expression of CSC plasticity markers through NFκB signaling pathway.

炎性肿瘤微环境(TME)是肿瘤促发过程的关键驱动因素。肿瘤相关巨噬细胞是肿瘤微环境中的主要免疫细胞类型之一,其密度的增加与前列腺癌的不良预后有关。在这里,我们研究了促炎性(M1)和免疫抑制性(M2)巨噬细胞对前列腺癌血统可塑性的影响。我们的研究结果表明,M1巨噬细胞分泌的因子会上调前列腺癌细胞中与干性相关的基因,同时下调与雄激素反应相关的基因。癌症干细胞(CSC)可塑性标志物NANOG、KLF4、SOX2、OCT4和CD44的表达受到M1巨噬细胞分泌因子的刺激。此外,在使用 M1 巨噬细胞分泌因子处理的 LNCaP 细胞中,还观察到 AR 及其靶基因 PSA 受到抑制。使用IKK16抑制剂抑制NFκB信号传导会导致NANOG、SOX2和CD44的下调以及CSC的可塑性。我们的研究强调,M1巨噬细胞分泌的因子通过NFκB信号通路上调CSC可塑性标志物的表达,从而驱动前列腺癌细胞的可塑性。
{"title":"Secreted factors from M1 macrophages drive prostate cancer stem cell plasticity by upregulating NANOG, <i>SOX2</i>, and <i>CD44</i> through NFκB-signaling.","authors":"Kirsi Kainulainen, Einari A Niskanen, Johanna Kinnunen, Kaisa Mäki-Mantila, Kiia Hartikainen, Ville Paakinaho, Marjo Malinen, Kirsi Ketola, Sanna Pasonen-Seppänen","doi":"10.1080/2162402X.2024.2393442","DOIUrl":"10.1080/2162402X.2024.2393442","url":null,"abstract":"<p><p>The inflammatory tumor microenvironment (TME) is a key driver for tumor-promoting processes. Tumor-associated macrophages are one of the main immune cell types in the TME and their increased density is related to poor prognosis in prostate cancer. Here, we investigated the influence of pro-inflammatory (M1) and immunosuppressive (M2) macrophages on prostate cancer lineage plasticity. Our findings reveal that M1 macrophage secreted factors upregulate genes related to stemness while downregulating genes associated with androgen response in prostate cancer cells. The expression of cancer stem cell (CSC) plasticity markers NANOG, KLF4, <i>SOX2, OCT4</i>, and CD44 was stimulated by the secreted factors from M1 macrophages. Moreover, AR and its target gene <i>PSA</i> were observed to be suppressed in LNCaP cells treated with secreted factors from M1 macrophages. Inhibition of NFκB signaling using the IKK16 inhibitor resulted in downregulation of NANOG, <i>SOX2</i>, and <i>CD44</i> and CSC plasticity. Our study highlights that the secreted factors from M1 macrophages drive prostate cancer cell plasticity by upregulating the expression of CSC plasticity markers through NFκB signaling pathway.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11340773/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142034397","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Oncoimmunology
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