Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A088
S. Majocchi, V. Moine, X. Chauchet, Lucile Broyer, L. Cons, L. Chatel, E. Hatterer, V. Buatois, H. Haddouk, Gérard Didelot, G. Magistrelli, Y. Poitevin, U. Ravn, A. Papaioannou, F. Richard, L. Shang, M. Kosco-Vilbois, N. Fischer, W. Ferlin, K. Masternak
Up-regulation of CD47 is an immune evasion mechanism used by different cancers to evade immune surveillance. Through its interaction with signal-regulatory protein alpha (SIRPα) on myeloid cells, CD47 delivers a universal “don’t eat me” signal to phagocytes, which prevents immune cells from efficiently eliminating tumor cells. Blockade of the SIRPα–CD47 innate immune checkpoint has therefore emerged as a new way to treat cancer. Several CD47-targeting molecules are in development with encouraging results obtained with monoclonal antibodies (mAb). However, the pharmacologic properties and the safety profile of molecules indiscriminately blocking CD47 can be improved by selectively inhibiting CD47 only on tumor cells. For this purpose, we generated bispecific antibodies (bsAbs) capable of targeting blockade of CD47 specifically to malignanT-cells through the co-engagement of a tumor-associated antigen (TAA). The bsAb NI-1801 specifically targets mesothelin (MSLN)-positive tumors. NI-1801 was shown to bind to MSLN-positive tumor cells, but not to MSLN-negative cells expressing physiologic levels of CD47 (e.g., leukocytes, erythrocytes, platelets). NI-1801 blocks the CD47-SIRPα interaction in a MSLN-dependent manner and thus minimizes the side effects related to a nonspecific blockade of CD47 on healthy cells. Studying antibody-dependent cellular phagocytosis (ADCP) and antibody-dependent cellular cytotoxicity (ADCC) of various MSLN-positive human tumor cell lines revealed that NI-1801 markedly enhanced killing as compared to amatuximab (an anti-MSLN mAb in clinical trials) and to the corresponding anti-MSLN mAb, exemplifying the role of blocking the “don’t eat me” signal to target cancer. NI-1801 also showed efficacy in various xenograft tumor models and analysis of the tumor microenvironment (TME) revealed a significant increase in leukocyte subpopulations (macrophages/monocytes and NK cells) of NI-1801 treated mice, suggesting that NI-1801 mediates the recruitment of monocytes from blood. Additionally, NI-1801 treatment affected the ratio between MHC-II-low and MHC-II-high macrophages in the TME. Finally, nonhuman primate studies with NI-1801 demonstrated a linear elimination profile, minimal target-mediated drug disposition and no hematologic toxicity. Taken together, these results illustrate that this strategy possesses potent anticancer activities both in vitro and in vivo in conjunction with favorable pharmacologic and toxicologic profiles. Citation Format: Stefano Majocchi, Valery Moine, Xavier Chauchet, Lucile Broyer, Laura Cons, Laurence Chatel, Eric Hatterer, Vanessa Buatois, Hasnaa Haddouk, Gerard Didelot, Giovanni Magistrelli, Yves Poitevin, Ulla Ravn, Anne Papaioannou, Francoise Richard, Limin Shang, Marie H. Kosco-Vilbois, Nicolas Fischer, Walter G. Ferlin, Krzysztof Masternak. Selective blockage of the innate immune checkpoint receptor CD47 on mesothelin (MSLN) positive solid tumor cells via dual targeting bispecific antibodies alters
{"title":"Abstract A088: Selective blockage of the innate immune checkpoint receptor CD47 on mesothelin (MSLN) positive solid tumor cells via dual targeting bispecific antibodies alters the tumor microenvironment to control tumor growth","authors":"S. Majocchi, V. Moine, X. Chauchet, Lucile Broyer, L. Cons, L. Chatel, E. Hatterer, V. Buatois, H. Haddouk, Gérard Didelot, G. Magistrelli, Y. Poitevin, U. Ravn, A. Papaioannou, F. Richard, L. Shang, M. Kosco-Vilbois, N. Fischer, W. Ferlin, K. Masternak","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A088","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A088","url":null,"abstract":"Up-regulation of CD47 is an immune evasion mechanism used by different cancers to evade immune surveillance. Through its interaction with signal-regulatory protein alpha (SIRPα) on myeloid cells, CD47 delivers a universal “don’t eat me” signal to phagocytes, which prevents immune cells from efficiently eliminating tumor cells. Blockade of the SIRPα–CD47 innate immune checkpoint has therefore emerged as a new way to treat cancer. Several CD47-targeting molecules are in development with encouraging results obtained with monoclonal antibodies (mAb). However, the pharmacologic properties and the safety profile of molecules indiscriminately blocking CD47 can be improved by selectively inhibiting CD47 only on tumor cells. For this purpose, we generated bispecific antibodies (bsAbs) capable of targeting blockade of CD47 specifically to malignanT-cells through the co-engagement of a tumor-associated antigen (TAA). The bsAb NI-1801 specifically targets mesothelin (MSLN)-positive tumors. NI-1801 was shown to bind to MSLN-positive tumor cells, but not to MSLN-negative cells expressing physiologic levels of CD47 (e.g., leukocytes, erythrocytes, platelets). NI-1801 blocks the CD47-SIRPα interaction in a MSLN-dependent manner and thus minimizes the side effects related to a nonspecific blockade of CD47 on healthy cells. Studying antibody-dependent cellular phagocytosis (ADCP) and antibody-dependent cellular cytotoxicity (ADCC) of various MSLN-positive human tumor cell lines revealed that NI-1801 markedly enhanced killing as compared to amatuximab (an anti-MSLN mAb in clinical trials) and to the corresponding anti-MSLN mAb, exemplifying the role of blocking the “don’t eat me” signal to target cancer. NI-1801 also showed efficacy in various xenograft tumor models and analysis of the tumor microenvironment (TME) revealed a significant increase in leukocyte subpopulations (macrophages/monocytes and NK cells) of NI-1801 treated mice, suggesting that NI-1801 mediates the recruitment of monocytes from blood. Additionally, NI-1801 treatment affected the ratio between MHC-II-low and MHC-II-high macrophages in the TME. Finally, nonhuman primate studies with NI-1801 demonstrated a linear elimination profile, minimal target-mediated drug disposition and no hematologic toxicity. Taken together, these results illustrate that this strategy possesses potent anticancer activities both in vitro and in vivo in conjunction with favorable pharmacologic and toxicologic profiles. Citation Format: Stefano Majocchi, Valery Moine, Xavier Chauchet, Lucile Broyer, Laura Cons, Laurence Chatel, Eric Hatterer, Vanessa Buatois, Hasnaa Haddouk, Gerard Didelot, Giovanni Magistrelli, Yves Poitevin, Ulla Ravn, Anne Papaioannou, Francoise Richard, Limin Shang, Marie H. Kosco-Vilbois, Nicolas Fischer, Walter G. Ferlin, Krzysztof Masternak. Selective blockage of the innate immune checkpoint receptor CD47 on mesothelin (MSLN) positive solid tumor cells via dual targeting bispecific antibodies alters","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"91205405","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A101
G. Prendergast, A. Nevler, A. Muller, E. Sutanto‐Ward, J. DuHadaway, K. Nagatomo, Eric Londin, K. O'Hayer, J. Cozzitorto, H. Lavu, T. Yeo, M. Curtis, Tatiana M. Villatoro, B. Leiby, J. Winter, C. Yeo, J. Brody
Sporadic pancreatic ductal adenocarcinoma (PDAC) develops into a lethal disease that has remained refractory to different treatment approaches including recent advances in cancer immunotherapy. Variations in host genetic status affecting the inflammatory microenvironment can impact cancer susceptibility, malignant progression and clinical outcomes. In this study, we present genetic evidence from mouse and human genetic studies supporting a role for IDO2, an immunometabolic modifier of B cell-mediated autoimmune responses, in promoting pancreatic ductal adenocarcinoma (PDAC). In an established transgenic mouse model of KRAS-induced pancreatic cancer, IDO2 genetic inactivation markedly reduced malignant progression. In retrospective clinical analyses of PDAC patients (N=200), the biallelic occurrence of either of two inactivating polymorphisms in the IDO2 coding region trended with favorable disease-free survival. In PDAC tissues, an inactive IDO2 host genotype corresponded with changes in expression of genes involved in tryptophan catabolism and immune modulation, along with a reduced neutrophil to lymphocyte ratio. Notably, subset analysis revealed a striking association of inactive IDO2 status with improved disease-free survival in patients who had received adjuvant radiotherapy, a treatment modality that has been highly debated due to its variable efficacy in patients. Accordingly, our findings suggest that host IDO2 genetic status may offer a simple incisive marker to stratify PDAC patients who stand to gain the most from adjuvant radiotherapy, addressing the long-standing debate of its benefits. Citation Format: George C. Prendergast, Avinoam Nevler, Alexander J. Muller, Erika Sutanto-Ward, James B. DuHadaway, Kei Nagatomo, Eric Londin, Kevin O9Hayer, Joseph A. Cozzitorto, Harish Lavu, Theresa P. Yeo, Mark Curtis, Tatiana Villatoro, Benjamin E. Leiby, Jordan M. Winter, Charles J. Yeo, Jonathan R. Brody. IDO2 host genetic status influences progression and radiotherapy response in pancreatic ductal adenocarcinoma [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A101.
散发性胰腺导管腺癌(PDAC)发展成为一种致死性疾病,对各种治疗方法(包括癌症免疫治疗的最新进展)仍然难治性。影响炎症微环境的宿主遗传状态的变化可以影响癌症易感性、恶性进展和临床结果。在这项研究中,我们提供了来自小鼠和人类遗传研究的遗传证据,支持IDO2 (B细胞介导的自身免疫反应的免疫代谢调节剂)在促进胰腺导管腺癌(PDAC)中的作用。在kras诱导的胰腺癌转基因小鼠模型中,IDO2基因失活显著降低了恶性进展。在PDAC患者(N=200)的回顾性临床分析中,IDO2编码区两种失活多态性中的任何一种双等位基因的出现都倾向于有利的无病生存。在PDAC组织中,失活的IDO2宿主基因型与色氨酸分解代谢和免疫调节相关基因的表达变化相对应,同时中性粒细胞与淋巴细胞比例降低。值得注意的是,亚群分析显示,在接受辅助放疗的患者中,IDO2不活跃状态与无病生存期的改善存在显著关联,辅助放疗是一种治疗方式,由于其对患者的疗效不一,一直备受争议。因此,我们的研究结果表明,宿主IDO2遗传状态可能提供一个简单而深刻的标记,以区分哪些PDAC患者从辅助放疗中获益最多,解决了长期以来关于其益处的争论。引文格式:George C. Prendergast, Avinoam Nevler, Alexander J. Muller, Erika Sutanto-Ward, James B. DuHadaway, Kei Nagatomo, Eric Londin, Kevin O9Hayer, Joseph A. Cozzitorto, Harish Lavu, Theresa P. Yeo, Mark Curtis, Tatiana Villatoro, Benjamin E. Leiby, Jordan M. Winter, Charles J. Yeo, Jonathan R. Brody。IDO2宿主遗传状态影响胰腺导管腺癌的进展和放疗反应[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志,2019;7(2增刊):摘要nr A101。
{"title":"Abstract A101: IDO2 host genetic status influences progression and radiotherapy response in pancreatic ductal adenocarcinoma","authors":"G. Prendergast, A. Nevler, A. Muller, E. Sutanto‐Ward, J. DuHadaway, K. Nagatomo, Eric Londin, K. O'Hayer, J. Cozzitorto, H. Lavu, T. Yeo, M. Curtis, Tatiana M. Villatoro, B. Leiby, J. Winter, C. Yeo, J. Brody","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A101","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A101","url":null,"abstract":"Sporadic pancreatic ductal adenocarcinoma (PDAC) develops into a lethal disease that has remained refractory to different treatment approaches including recent advances in cancer immunotherapy. Variations in host genetic status affecting the inflammatory microenvironment can impact cancer susceptibility, malignant progression and clinical outcomes. In this study, we present genetic evidence from mouse and human genetic studies supporting a role for IDO2, an immunometabolic modifier of B cell-mediated autoimmune responses, in promoting pancreatic ductal adenocarcinoma (PDAC). In an established transgenic mouse model of KRAS-induced pancreatic cancer, IDO2 genetic inactivation markedly reduced malignant progression. In retrospective clinical analyses of PDAC patients (N=200), the biallelic occurrence of either of two inactivating polymorphisms in the IDO2 coding region trended with favorable disease-free survival. In PDAC tissues, an inactive IDO2 host genotype corresponded with changes in expression of genes involved in tryptophan catabolism and immune modulation, along with a reduced neutrophil to lymphocyte ratio. Notably, subset analysis revealed a striking association of inactive IDO2 status with improved disease-free survival in patients who had received adjuvant radiotherapy, a treatment modality that has been highly debated due to its variable efficacy in patients. Accordingly, our findings suggest that host IDO2 genetic status may offer a simple incisive marker to stratify PDAC patients who stand to gain the most from adjuvant radiotherapy, addressing the long-standing debate of its benefits. Citation Format: George C. Prendergast, Avinoam Nevler, Alexander J. Muller, Erika Sutanto-Ward, James B. DuHadaway, Kei Nagatomo, Eric Londin, Kevin O9Hayer, Joseph A. Cozzitorto, Harish Lavu, Theresa P. Yeo, Mark Curtis, Tatiana Villatoro, Benjamin E. Leiby, Jordan M. Winter, Charles J. Yeo, Jonathan R. Brody. IDO2 host genetic status influences progression and radiotherapy response in pancreatic ductal adenocarcinoma [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A101.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"90395981","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A087
A. Li, C. J. Cao, G. C. Chan
Mesenchymal stem cells (MSCs) are a heterogeneous population of multipotenT-cells and are capable of differentiating into certain cell types. They can be found within tumor core and constitute part of the tumor microenvironment. Their role in tumor progression remains contradictory with both sides arguing pro- and anti-tumor function. In regarding to the pro-tumor effect, MSCs have shown to have immunoregulatory properties—suppressing T lymphocytes, B lymphocytes and dendritic cells, thereby helping tumor cells to escape from the immune surveillance. However there are some studies that showed certain microenvironment and stimulation can drive MSC into a proinflammatory phenotype. During cancer therapy, large amount of apoptotic cells are released to the tumor microenvironment. The aim of this study is to investigate how an apoptotic microenvironment can affect the function of MSC on naive T-cells. Apoptotic cells (ACs) were derived from the neuroblastoma cell line SK-N-LP treated with cisplatin for 24 hr; they were then co-cultured with the immortalised human MSC cell line (hTMSC). Naive T-cells were isolated from peripheral blood mononuclear cells of healthy donors and further co-cultured with either hTMSC alone or AC-treated hTMSC (hTMSC-AC) for 5 days. Cell proliferation and phenotype were analyzed by flow cytometry. We demonstrated that hTMSCs have the ability to phagocytose AC. After engulfment, the hTMSC-AC were able to increase T-cell proliferation compared to those co-cultured with untreated hTMSC. The T-cells co-cultured with hTMSC-AC also showed a decrease in CD45RA expression, suggesting an increase in T-cell activation. Moreover, these T-cells had a higher expression level of interferon-gamma (IFN-γ) and a lower expression of FoxP3, indicating a shift towards proinflammatory effector T-cells and a reduction of regulatory T-cells. Consistent with other literature, hTMSC has the ability to supress T-cells’ activation and proliferation. However in the presence of AC, hTMSC immunosuppressive property was impeded, thereby heightening the immune response. Therefore the microenvironment can play a major factor in determining the role of MSC on tumor. Further investigation of the interaction of MSC with immune cells in an apoptotic tumor microenvironment can lead to a potential immunotherapy for neuroblastoma. Citation Format: Anita K.Y. Li, Carmen J.M. Cao, Godfrey C.F. Chan. The immunosuppressive property of mesenchymal stem cell in an apoptotic tumor microenvironment [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A087.
{"title":"Abstract A087: The immunosuppressive property of mesenchymal stem cell in an apoptotic tumor microenvironment","authors":"A. Li, C. J. Cao, G. C. Chan","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A087","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A087","url":null,"abstract":"Mesenchymal stem cells (MSCs) are a heterogeneous population of multipotenT-cells and are capable of differentiating into certain cell types. They can be found within tumor core and constitute part of the tumor microenvironment. Their role in tumor progression remains contradictory with both sides arguing pro- and anti-tumor function. In regarding to the pro-tumor effect, MSCs have shown to have immunoregulatory properties—suppressing T lymphocytes, B lymphocytes and dendritic cells, thereby helping tumor cells to escape from the immune surveillance. However there are some studies that showed certain microenvironment and stimulation can drive MSC into a proinflammatory phenotype. During cancer therapy, large amount of apoptotic cells are released to the tumor microenvironment. The aim of this study is to investigate how an apoptotic microenvironment can affect the function of MSC on naive T-cells. Apoptotic cells (ACs) were derived from the neuroblastoma cell line SK-N-LP treated with cisplatin for 24 hr; they were then co-cultured with the immortalised human MSC cell line (hTMSC). Naive T-cells were isolated from peripheral blood mononuclear cells of healthy donors and further co-cultured with either hTMSC alone or AC-treated hTMSC (hTMSC-AC) for 5 days. Cell proliferation and phenotype were analyzed by flow cytometry. We demonstrated that hTMSCs have the ability to phagocytose AC. After engulfment, the hTMSC-AC were able to increase T-cell proliferation compared to those co-cultured with untreated hTMSC. The T-cells co-cultured with hTMSC-AC also showed a decrease in CD45RA expression, suggesting an increase in T-cell activation. Moreover, these T-cells had a higher expression level of interferon-gamma (IFN-γ) and a lower expression of FoxP3, indicating a shift towards proinflammatory effector T-cells and a reduction of regulatory T-cells. Consistent with other literature, hTMSC has the ability to supress T-cells’ activation and proliferation. However in the presence of AC, hTMSC immunosuppressive property was impeded, thereby heightening the immune response. Therefore the microenvironment can play a major factor in determining the role of MSC on tumor. Further investigation of the interaction of MSC with immune cells in an apoptotic tumor microenvironment can lead to a potential immunotherapy for neuroblastoma. Citation Format: Anita K.Y. Li, Carmen J.M. Cao, Godfrey C.F. Chan. The immunosuppressive property of mesenchymal stem cell in an apoptotic tumor microenvironment [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A087.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"84530801","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A084
J. Krolewski, K. Sha, M. Mastri, D. Tang, K. Eng, K. Nastiuk
Most prostate cancer (PCa) deaths are due to castration-resistant PCa (CRPC), following failure of androgen-deprivation therapy (ADT). ADT is the standard of care for patients with advanced PCa. However, nearly universal progression to castration-resistant prostate cancer (CRPC) occurs 2-3 years after ADT is initiated. Although there have been recent improvements in the treatment of CRPC, even the most promising therapies are still not curative. One approach to this problem is to improve the initial treatment of advanced prostate cancers, by combining complementary therapies with ADT, to prevent progression of such advanced cancers to CRPC. Immunotherapy with checkpoint inhibitors (CPIs) has not been effective in prostate cancers, perhaps because such cancers are “cold” (lacking cytolytic CD8 T-cells). Some cold tumors may be caused by infiltration of myeloid cell populations (tumor associated macrophages and myeloid-derived suppressor cells) into the tumor immune cell microenvironment (TIME). Recently, we found that in a PTEN-deficient mouse PCa model, castration induces an immunosuppressive state within the tumor that is concurrent with tumor recurrence. The response to castration/ADT is tri-phasic: a pro-apoptotic regression phase when tumor shrinks, followed by selection for a residual population of resistant tumor cells and finally recurrent growth as CRPC. Using PCa cell lines to model the first two phases of the response to ADT, we have shown that ADT induces apoptosis, thereby enriching for an ADT-resistant stem/progenitor population that we propose is the in vivo source of TNF. Mechanistically, in our model system the response to ADT is driven by the soluble mediators TNF and CCL2, which facilitate communication within the TIME. Specifically, a TNF-CCL2-CCR2 paracrine loop is induced between prostate cancer cells and non-tumor cells in the microenvironment: TNF produced by tumor cells acts on myofibroblasts to induce CCL2 production, which in turn recruits CCR2+ tumor-associated macrophages (TAMs). To investigate the ADT response within the TIME in an in vivo model of prostate cancer, we employed a prostate-specific PTEN-deficient mouse model (PbCre4 x PTENf/f). Castration caused the tumors to regress, consistent with initial phase of the response that is seen in the human disease. At late times post-castration (5-6 weeks), corresponding to the selection phase, we observed a coordinate increase in the stem/progenitor tumor cell population, as well as TNF and CCL2, within the TIME. Immunohistochemical staining of tumors 5 weeks post-castration revealed an increase in TAMs, and a decrease in CD8 T-cells, consistent with an immunosuppressive or immunoevasive state. This phenotype was reversed by a soluble receptor that binds TNF (etanercept). We also observed increased myeloid-derived suppressor cells (MDSC). Thus, following ADT, TNF derived from an ADT-resistant stem/progenitor epithelial tumor cell population promotes an immunosuppressive
大多数前列腺癌(PCa)死亡是由于雄激素剥夺治疗(ADT)失败后的去势抵抗性前列腺癌(CRPC)。ADT是晚期前列腺癌患者的标准治疗方法。然而,几乎普遍进展为去势抵抗性前列腺癌(CRPC)发生在ADT开始后2-3年。尽管最近CRPC的治疗有了进步,但即使是最有希望的治疗方法仍然无法治愈。解决这一问题的一种方法是通过将补充疗法与ADT相结合来改善晚期前列腺癌的初始治疗,以防止这种晚期癌症进展为CRPC。使用检查点抑制剂(CPIs)的免疫疗法对前列腺癌没有效果,可能是因为这种癌症是“冷的”(缺乏细胞溶解性CD8 t细胞)。一些冷肿瘤可能是由骨髓细胞群(肿瘤相关巨噬细胞和骨髓源性抑制细胞)浸润到肿瘤免疫细胞微环境(TIME)引起的。最近,我们发现在pten缺失的小鼠PCa模型中,阉割诱导肿瘤内的免疫抑制状态,并伴有肿瘤复发。对去势/ADT的反应是三个阶段:肿瘤缩小时的促凋亡消退阶段,随后选择残余的耐药肿瘤细胞群,最后作为CRPC复发生长。利用PCa细胞系模拟ADT反应的前两个阶段,我们已经证明ADT诱导细胞凋亡,从而丰富ADT抗性干细胞/祖细胞群,我们认为这是TNF的体内来源。从机制上讲,在我们的模型系统中,对ADT的反应是由可溶性介质TNF和CCL2驱动的,它们促进了TIME内的通信。具体来说,微环境中,前列腺癌细胞和非肿瘤细胞之间诱导了一个TNF-CCL2-CCR2旁分泌环:肿瘤细胞产生的TNF作用于肌成纤维细胞诱导CCL2的产生,进而招募CCR2+肿瘤相关巨噬细胞(tam)。为了在前列腺癌体内模型中研究ADT在时间内的反应,我们采用了前列腺特异性pten缺陷小鼠模型(PbCre4 x pten /f)。去势导致肿瘤消退,与人类疾病反应的初始阶段一致。在去势后的后期(5-6周),与选择阶段相对应,我们观察到在时间内干细胞/祖细胞群以及TNF和CCL2的坐标增加。阉割后5周的肿瘤免疫组化染色显示tam增加,CD8 t细胞减少,符合免疫抑制或免疫逃避状态。这种表型被结合TNF(依那西普)的可溶性受体逆转。我们还观察到髓源性抑制细胞(MDSC)的增加。因此,在ADT后,来自ADT耐药干细胞/祖上皮肿瘤细胞群的TNF在TIME中通过CCL2促进免疫抑制状态。对公开的人PCa数据集的分析显示,肿瘤坏死因子和干细胞/祖细胞标志物在CRPC中的表达均增加,这与我们的假设一致,即ADT通过TNF- ccl2 - ccr2轴驱动免疫抑制状态的发展。我们的研究结果为未来开发能够提高ADT疗效的免疫疗法奠定了基础。引用格式:John J. Krolewski, Kai Sha, Michalis Mastri, Dean Tang, Kevin Eng, Kent L. Nastiuk。雄激素剥夺联合免疫治疗预防去势抵抗性前列腺癌进展的研究[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A084。
{"title":"Abstract A084: Towards combining androgen deprivation and immunotherapy to prevent progression to castration-resistant prostate cancer","authors":"J. Krolewski, K. Sha, M. Mastri, D. Tang, K. Eng, K. Nastiuk","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A084","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A084","url":null,"abstract":"Most prostate cancer (PCa) deaths are due to castration-resistant PCa (CRPC), following failure of androgen-deprivation therapy (ADT). ADT is the standard of care for patients with advanced PCa. However, nearly universal progression to castration-resistant prostate cancer (CRPC) occurs 2-3 years after ADT is initiated. Although there have been recent improvements in the treatment of CRPC, even the most promising therapies are still not curative. One approach to this problem is to improve the initial treatment of advanced prostate cancers, by combining complementary therapies with ADT, to prevent progression of such advanced cancers to CRPC. Immunotherapy with checkpoint inhibitors (CPIs) has not been effective in prostate cancers, perhaps because such cancers are “cold” (lacking cytolytic CD8 T-cells). Some cold tumors may be caused by infiltration of myeloid cell populations (tumor associated macrophages and myeloid-derived suppressor cells) into the tumor immune cell microenvironment (TIME). Recently, we found that in a PTEN-deficient mouse PCa model, castration induces an immunosuppressive state within the tumor that is concurrent with tumor recurrence. The response to castration/ADT is tri-phasic: a pro-apoptotic regression phase when tumor shrinks, followed by selection for a residual population of resistant tumor cells and finally recurrent growth as CRPC. Using PCa cell lines to model the first two phases of the response to ADT, we have shown that ADT induces apoptosis, thereby enriching for an ADT-resistant stem/progenitor population that we propose is the in vivo source of TNF. Mechanistically, in our model system the response to ADT is driven by the soluble mediators TNF and CCL2, which facilitate communication within the TIME. Specifically, a TNF-CCL2-CCR2 paracrine loop is induced between prostate cancer cells and non-tumor cells in the microenvironment: TNF produced by tumor cells acts on myofibroblasts to induce CCL2 production, which in turn recruits CCR2+ tumor-associated macrophages (TAMs). To investigate the ADT response within the TIME in an in vivo model of prostate cancer, we employed a prostate-specific PTEN-deficient mouse model (PbCre4 x PTENf/f). Castration caused the tumors to regress, consistent with initial phase of the response that is seen in the human disease. At late times post-castration (5-6 weeks), corresponding to the selection phase, we observed a coordinate increase in the stem/progenitor tumor cell population, as well as TNF and CCL2, within the TIME. Immunohistochemical staining of tumors 5 weeks post-castration revealed an increase in TAMs, and a decrease in CD8 T-cells, consistent with an immunosuppressive or immunoevasive state. This phenotype was reversed by a soluble receptor that binds TNF (etanercept). We also observed increased myeloid-derived suppressor cells (MDSC). Thus, following ADT, TNF derived from an ADT-resistant stem/progenitor epithelial tumor cell population promotes an immunosuppressive","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"85988936","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A111
Michael Schulz, Katja Niesel, Anna Salamero Boix, Woongjoo Chae, Birgitta E. Michels, A. Schaeffer, Maja I. Strecker, T. Alekseeva, S. Stein, H. Farin, F. Roedel, P. Harter, K. Plate, L. Sevenich
Brain metastases represent the most common intracranial tumor in adults associated with poor prognosis and median survival of only a few months. Despite current success in the development of targeted or immuno-therapies against different cancer entities, those strategies are ineffective against brain metastases. Hence, treatment options for brain metastasis patients largely remain limited to surgical resection and radio- and/or chemotherapy. This paucity can in part be attributed to the immune-privileged status of the brain where the blood brain-barrier restricts the entry of blood-borne immune cells. However, recent insights into the immune landscape of primary brain cancers indicate that tumor progression leads to an infiltration of blood-borne immune cells into the brain. We employ a comprehensive set of experimental brain metastasis models to characterize the immune landscape of brain metastases from different primary cancer entities at distinct disease stages and in response to radiotherapy. Our data indicate that brain metastases induce massive infiltration of myeloid and lymphoid cell populations into the central nervous system. This leads to the establishment of a dynamic and highly complex tumor microenvironment that affects tumor progression and therapy response. Fractionated whole-brain radiotherapy leads to enhanced infiltration of blood-borne myeloid and lymphoid cells. Transcriptome analysis of brain-resident and recruited myeloid cells indicate a switch from a proinflammatory towards an immune-suppressive environment at advanced disease stages. Importantly, radiotherapy was found to induce gene signatures that are associated with proinflammatory innate immune responses that could revert the establishment of an immune-suppressive environment. Consequently, radiotherapy might sensitize brain metastases towards immuno-therapies. Our goal is to identify pathways or molecular targets that are induced by radiotherapy in the tumor microenvironment to overcome resistance against immuno-therapy. In this project, we seek to test strategies to maintain or induce proinflammatory immune responses for improved targeted or immuno-therapies against brain metastasis. Citation Format: Michael Schulz, Katja Niesel, Anna Salamero Boix, Woon Hyung Chae, Birgitta Michels, Alexander Schaeffer, Maja Strecker, Tijna Alekseeva, Stefan Stein, Henner Farin, Franz Roedel, Patrick Harter, Karlheinz Plate, and Lisa Sevenich. Effects of ionizing radiation on brain metastasis-associated inflammation and its implication for immunotherapy [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A111.
脑转移瘤是成人最常见的颅内肿瘤,预后差,中位生存期只有几个月。尽管目前在针对不同癌症实体的靶向或免疫治疗方面取得了成功,但这些策略对脑转移无效。因此,脑转移患者的治疗选择很大程度上仍然局限于手术切除和放疗和/或化疗。这种缺乏可以部分归因于大脑的免疫特权地位,其中血脑屏障限制了血源性免疫细胞的进入。然而,最近对原发性脑癌免疫景观的研究表明,肿瘤进展导致血源性免疫细胞渗入大脑。我们采用了一套全面的实验性脑转移模型来表征不同原发性肿瘤实体在不同疾病阶段和对放疗的反应的脑转移的免疫景观。我们的数据表明,脑转移诱导髓细胞和淋巴细胞群大量浸润到中枢神经系统。这导致建立一个动态和高度复杂的肿瘤微环境,影响肿瘤的进展和治疗反应。分块全脑放疗可增强血源性髓细胞和淋巴细胞的浸润。脑驻留和募集骨髓细胞的转录组分析表明,在疾病晚期,从促炎环境向免疫抑制环境转变。重要的是,研究发现放射治疗可以诱导与促炎先天免疫反应相关的基因特征,从而恢复免疫抑制环境的建立。因此,放射治疗可能使脑转移灶对免疫治疗敏感。我们的目标是确定肿瘤微环境中放疗诱导的途径或分子靶点,以克服对免疫治疗的抵抗。在这个项目中,我们试图测试维持或诱导促炎免疫反应的策略,以改善针对脑转移的靶向或免疫治疗。引文格式:Michael Schulz, Katja Niesel, Anna Salamero Boix, Woon Hyung Chae, Birgitta Michels, Alexander Schaeffer, Maja Strecker, Tijna Alekseeva, Stefan Stein, Henner Farin, Franz Roedel, Patrick Harter, Karlheinz Plate和Lisa Sevenich。电离辐射对脑转移相关炎症的影响及其对免疫治疗的启示[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫,2019;7(2增刊):摘要nr A111。
{"title":"Abstract A111: Effects of ionizing radiation on brain metastasis-associated inflammation and its implication for immunotherapy","authors":"Michael Schulz, Katja Niesel, Anna Salamero Boix, Woongjoo Chae, Birgitta E. Michels, A. Schaeffer, Maja I. Strecker, T. Alekseeva, S. Stein, H. Farin, F. Roedel, P. Harter, K. Plate, L. Sevenich","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A111","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A111","url":null,"abstract":"Brain metastases represent the most common intracranial tumor in adults associated with poor prognosis and median survival of only a few months. Despite current success in the development of targeted or immuno-therapies against different cancer entities, those strategies are ineffective against brain metastases. Hence, treatment options for brain metastasis patients largely remain limited to surgical resection and radio- and/or chemotherapy. This paucity can in part be attributed to the immune-privileged status of the brain where the blood brain-barrier restricts the entry of blood-borne immune cells. However, recent insights into the immune landscape of primary brain cancers indicate that tumor progression leads to an infiltration of blood-borne immune cells into the brain. We employ a comprehensive set of experimental brain metastasis models to characterize the immune landscape of brain metastases from different primary cancer entities at distinct disease stages and in response to radiotherapy. Our data indicate that brain metastases induce massive infiltration of myeloid and lymphoid cell populations into the central nervous system. This leads to the establishment of a dynamic and highly complex tumor microenvironment that affects tumor progression and therapy response. Fractionated whole-brain radiotherapy leads to enhanced infiltration of blood-borne myeloid and lymphoid cells. Transcriptome analysis of brain-resident and recruited myeloid cells indicate a switch from a proinflammatory towards an immune-suppressive environment at advanced disease stages. Importantly, radiotherapy was found to induce gene signatures that are associated with proinflammatory innate immune responses that could revert the establishment of an immune-suppressive environment. Consequently, radiotherapy might sensitize brain metastases towards immuno-therapies. Our goal is to identify pathways or molecular targets that are induced by radiotherapy in the tumor microenvironment to overcome resistance against immuno-therapy. In this project, we seek to test strategies to maintain or induce proinflammatory immune responses for improved targeted or immuno-therapies against brain metastasis. Citation Format: Michael Schulz, Katja Niesel, Anna Salamero Boix, Woon Hyung Chae, Birgitta Michels, Alexander Schaeffer, Maja Strecker, Tijna Alekseeva, Stefan Stein, Henner Farin, Franz Roedel, Patrick Harter, Karlheinz Plate, and Lisa Sevenich. Effects of ionizing radiation on brain metastasis-associated inflammation and its implication for immunotherapy [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A111.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87051765","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A119
J. Wenthe, Mantas Šilanskas, E. Eriksson, A. Loskog
In recent years, cancer immunotherapies significantly advanced the clinical management of metastatic melanoma. In particular, treatment with immune checkpoint inhibitors increased survival rates compared with standard therapy. However, not all patients benefit from checkpoint blockade. Primary treatment resistance is connected to poor T-cell infiltration in the tumors. This can be due to limited activation of antigen-presentation cells such as dendritic cells (DCs) and the high threshold of activating low-affinity T-cells that may respond to the tumor antigens. CD27 signaling in T-cells during activation lowers the T-cell receptor signaling threshold, which may be important to activate tumor-targeting T-cells. Secondary resistance to checkpoint blockade therapy includes loss of MHC-I on the tumor cells. Hence, inducing natural killer (NK) cell activation in parallel to antitumor T-cells may be crucial. LOAd703 was designed to optimize antitumor immune activation. LOAd703 is an oncolytic adenovirus (serotype Ad5/35) carrying two immunostimulatory transgenes; a trimerized membrane-bound CD40 ligand (TMZ-CD40L) and the full-length 4-1BB ligand (4-1BBL) (patent filed: PCT/EP2015/057489). The viral replication is restricted to tumor cells with a hyperphosphorylated retinoblastoma pathway due to a deletion in E1A, but transgenes are expressed under the control of a cytomegalovirus (CMV) promoter, which enables transgene expression even in the surrounding tumor microenvironment. Herein, we investigated LOAd703 in a melanoma model and its immunostimulatory effect on DC maturation to induce antigen-specific T-cell responses. LOAd703 infected the human melanoma cell line 526-mel and efficiently induced tumor cell death as evaluated by MTS viability assay. The viability of infected cells was reduced to 15% at 72 hours post infection compared to uninfected cells. Transgene expression of both TMZ-CD40L and 4-1BBL was confirmed by flow cytometry post infection. To evaluate the immunostimulatory capacity of LOAd703, immature DCs were differentiated from CD14+ monocytes using granulocyte-macrophage colony-stimulating factor and interleukin-4 and infected with virus. LOAd703-infected DCs upregulated the expression of maturation markers, such as CD83, CD86 and MHC molecules as analyzed by flow cytometry. Interestingly, CD70 that is required for CD27 stimuli of T-cells was highly upregulated on the DCs using LOAd703. Furthermore, the chemokine receptor CCR7 and the adhesion molecule ICAM-1 were increased upon LOAd703 infection, which are crucial for lymph node homing and the initiation of a systemic response. Next, the functional capacity of LOAd703-matured DCs to induce antigen-specific T-cell responses was assessed in a CMV model, in which CMV-peptide pulsed DCs are utilized to induce expansion of CMV-specific T-cells. LOAd703-matured DCs from CMV+ donors were pulsed with CMV-peptide and co-cultured with autologous peripheral blood mononuclear cells for 11 days.
{"title":"Abstract A119: Combating primary and secondary checkpoint blockade resistance using immunostimulatory CD40L/4-1BBL-encoding oncolytic virotherapy for melanoma","authors":"J. Wenthe, Mantas Šilanskas, E. Eriksson, A. Loskog","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A119","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A119","url":null,"abstract":"In recent years, cancer immunotherapies significantly advanced the clinical management of metastatic melanoma. In particular, treatment with immune checkpoint inhibitors increased survival rates compared with standard therapy. However, not all patients benefit from checkpoint blockade. Primary treatment resistance is connected to poor T-cell infiltration in the tumors. This can be due to limited activation of antigen-presentation cells such as dendritic cells (DCs) and the high threshold of activating low-affinity T-cells that may respond to the tumor antigens. CD27 signaling in T-cells during activation lowers the T-cell receptor signaling threshold, which may be important to activate tumor-targeting T-cells. Secondary resistance to checkpoint blockade therapy includes loss of MHC-I on the tumor cells. Hence, inducing natural killer (NK) cell activation in parallel to antitumor T-cells may be crucial. LOAd703 was designed to optimize antitumor immune activation. LOAd703 is an oncolytic adenovirus (serotype Ad5/35) carrying two immunostimulatory transgenes; a trimerized membrane-bound CD40 ligand (TMZ-CD40L) and the full-length 4-1BB ligand (4-1BBL) (patent filed: PCT/EP2015/057489). The viral replication is restricted to tumor cells with a hyperphosphorylated retinoblastoma pathway due to a deletion in E1A, but transgenes are expressed under the control of a cytomegalovirus (CMV) promoter, which enables transgene expression even in the surrounding tumor microenvironment. Herein, we investigated LOAd703 in a melanoma model and its immunostimulatory effect on DC maturation to induce antigen-specific T-cell responses. LOAd703 infected the human melanoma cell line 526-mel and efficiently induced tumor cell death as evaluated by MTS viability assay. The viability of infected cells was reduced to 15% at 72 hours post infection compared to uninfected cells. Transgene expression of both TMZ-CD40L and 4-1BBL was confirmed by flow cytometry post infection. To evaluate the immunostimulatory capacity of LOAd703, immature DCs were differentiated from CD14+ monocytes using granulocyte-macrophage colony-stimulating factor and interleukin-4 and infected with virus. LOAd703-infected DCs upregulated the expression of maturation markers, such as CD83, CD86 and MHC molecules as analyzed by flow cytometry. Interestingly, CD70 that is required for CD27 stimuli of T-cells was highly upregulated on the DCs using LOAd703. Furthermore, the chemokine receptor CCR7 and the adhesion molecule ICAM-1 were increased upon LOAd703 infection, which are crucial for lymph node homing and the initiation of a systemic response. Next, the functional capacity of LOAd703-matured DCs to induce antigen-specific T-cell responses was assessed in a CMV model, in which CMV-peptide pulsed DCs are utilized to induce expansion of CMV-specific T-cells. LOAd703-matured DCs from CMV+ donors were pulsed with CMV-peptide and co-cultured with autologous peripheral blood mononuclear cells for 11 days. ","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"84024287","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A063
N. Miranda, N. D. Vries, V. V. Unen, T. Abdelaal, M. Ijsselsteijn, R. V. D. Breggen, A. Fariña-Sarasqueta, K. Peeters, T. Höllt, B. Lelieveldt, F. Koning
Checkpoint blockade has revived the potential of immunotherapy for cancer treatment. For optimal application and development of cancer immunotherapies, a comprehensive understanding of the antitumor immune response is required. We unraveled local and systemic immune profiles of colorectal cancer by multidimensional mass cytometric analysis of 36 immune cell markers at the single-cell level in tumor tissues, tumor-associated lymph nodes, adjacent normal mucosa, and peripheral blood samples from CRC patients. We identified 218 phenotypically distinct immune cell clusters, including a previously neglected innate lymphoid cell (CD7+CD3-CD127-CD45RO+CD56+) population with cytotoxic potential. This subset demonstrated a tissue-resident (CD69+, CD103+) phenotype, and was most abundant in the immunogenic mismatch repair deficient (MMRd) cancers. Furthermore, tumor-resident immune cell populations were identified across the adaptive (CD4+ and CD8+) and innate (gammadelta) T-cell compartments showing a highly similar activated (HLA-DR+, CD38+, PD-1+) phenotype. PD-1 intermediate and PD-1 high CD8+ T-cell subsets represented distinct states of T-cell activation that further discriminated immunogenic from non-immunogenic colorectal cancers. Remarkably, activated gammadelta T-cells were specific for MMRd cancers, and their potential role in the response to PD-1 checkpoint blockade requires further clarification. The nonactivated counterparts of the tumor-resident CD103+PD-1+ cytotoxic and gammadelta T-cells were present in both tumor and healthy colorectal tissues. We did not detect any of the aforementioned tumor-resident immune cell populations in lymph node samples, with the exception of a tumor-positive lymph node. This indicates that the critical immune cell populations with antitumor activity reside in the colorectal mucosa, and that the role of lymph nodes in the antitumor immune response should be revisited. Finally, by applying imaging mass cytometry we demonstrated that the cytotoxic anti-tumor response in colorectal cancer is highly diverse and not restricted to cytotoxic T-cells, which opens new avenues for the management of this disease.The findings presented here advance the paradigm of antitumor immunity in colorectal cancer and provide a blueprint for the detailed characterization of the involved immune cell subsets. The coordinated action of innate and adaptive immune cell populations suggests a multitargeted exploitation of their antitumor properties in a therapeutic setting. Citation Format: Noel F. de Miranda, Natasja L. de Vries, Vincent van Unen, Tamim Abdelaal, Marieke E. Ijsselsteijn, Ruud van der Breggen, Arantza Farina-Sarasqueta, Koen C.M.J. Peeters, Thomas Hollt, Boudewijn P.F. Lelieveldt, Frits Koning. Multidimensional cytometric analysis of colorectal cancer reveals novel and diverse mediators of antitumor immunity [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translat
检查点阻断已经恢复了癌症治疗免疫疗法的潜力。为了优化癌症免疫疗法的应用和发展,需要对抗肿瘤免疫反应有一个全面的了解。我们通过对结直肠癌患者肿瘤组织、肿瘤相关淋巴结、邻近正常粘膜和外周血样本中36种单细胞水平免疫细胞标记物的多维质量细胞分析,揭示了结直肠癌的局部和全身免疫特征。我们确定了218个表型不同的免疫细胞簇,包括以前被忽视的具有细胞毒性潜力的先天淋巴样细胞(CD7+CD3-CD127-CD45RO+CD56+)群体。该亚群表现出组织常驻(CD69+, CD103+)表型,并且在免疫原性错配修复缺陷(MMRd)癌症中最为丰富。此外,在适应性(CD4+和CD8+)和先天(γ - δ) t细胞区室中鉴定了肿瘤驻留免疫细胞群,显示出高度相似的活化(HLA-DR+, CD38+, PD-1+)表型。PD-1中间和PD-1高CD8+ t细胞亚群代表了不同的t细胞激活状态,进一步区分了免疫原性和非免疫原性结直肠癌。值得注意的是,活化的γ - δ t细胞对MMRd癌症是特异性的,它们在PD-1检查点阻断反应中的潜在作用需要进一步澄清。肿瘤组织和健康结肠组织中均存在肿瘤驻留CD103+PD-1+细胞毒性和γ - δ t细胞的非激活对应物。除了肿瘤阳性淋巴结外,我们没有在淋巴结样本中检测到任何上述肿瘤驻留免疫细胞群。这表明具有抗肿瘤活性的关键免疫细胞群存在于结直肠粘膜,并且淋巴结在抗肿瘤免疫应答中的作用应该被重新审视。最后,通过应用成像细胞计数技术,我们证明了结直肠癌的细胞毒性抗肿瘤反应是高度多样化的,并不局限于细胞毒性t细胞,这为这种疾病的治疗开辟了新的途径。本文提出的研究结果推进了结直肠癌抗肿瘤免疫的范式,并为相关免疫细胞亚群的详细表征提供了蓝图。先天免疫细胞群和适应性免疫细胞群的协同作用表明,在治疗环境中,它们的抗肿瘤特性是一个多目标的开发。引用格式:Noel F. de Miranda, Natasja L. de Vries, Vincent van Unen, Tamim Abdelaal, Marieke E. Ijsselsteijn, Ruud van der Breggen, Arantza Farina-Sarasqueta, Koen C.M.J. Peeters, Thomas Hollt, Boudewijn P.F. Lelieveldt, Frits Koning。结直肠癌的多维细胞分析揭示了新的多种抗肿瘤免疫介质[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A063。
{"title":"Abstract A063: Multidimensional cytometric analysis of colorectal cancer reveals novel and diverse mediators of antitumor immunity","authors":"N. Miranda, N. D. Vries, V. V. Unen, T. Abdelaal, M. Ijsselsteijn, R. V. D. Breggen, A. Fariña-Sarasqueta, K. Peeters, T. Höllt, B. Lelieveldt, F. Koning","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A063","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A063","url":null,"abstract":"Checkpoint blockade has revived the potential of immunotherapy for cancer treatment. For optimal application and development of cancer immunotherapies, a comprehensive understanding of the antitumor immune response is required. We unraveled local and systemic immune profiles of colorectal cancer by multidimensional mass cytometric analysis of 36 immune cell markers at the single-cell level in tumor tissues, tumor-associated lymph nodes, adjacent normal mucosa, and peripheral blood samples from CRC patients. We identified 218 phenotypically distinct immune cell clusters, including a previously neglected innate lymphoid cell (CD7+CD3-CD127-CD45RO+CD56+) population with cytotoxic potential. This subset demonstrated a tissue-resident (CD69+, CD103+) phenotype, and was most abundant in the immunogenic mismatch repair deficient (MMRd) cancers. Furthermore, tumor-resident immune cell populations were identified across the adaptive (CD4+ and CD8+) and innate (gammadelta) T-cell compartments showing a highly similar activated (HLA-DR+, CD38+, PD-1+) phenotype. PD-1 intermediate and PD-1 high CD8+ T-cell subsets represented distinct states of T-cell activation that further discriminated immunogenic from non-immunogenic colorectal cancers. Remarkably, activated gammadelta T-cells were specific for MMRd cancers, and their potential role in the response to PD-1 checkpoint blockade requires further clarification. The nonactivated counterparts of the tumor-resident CD103+PD-1+ cytotoxic and gammadelta T-cells were present in both tumor and healthy colorectal tissues. We did not detect any of the aforementioned tumor-resident immune cell populations in lymph node samples, with the exception of a tumor-positive lymph node. This indicates that the critical immune cell populations with antitumor activity reside in the colorectal mucosa, and that the role of lymph nodes in the antitumor immune response should be revisited. Finally, by applying imaging mass cytometry we demonstrated that the cytotoxic anti-tumor response in colorectal cancer is highly diverse and not restricted to cytotoxic T-cells, which opens new avenues for the management of this disease.The findings presented here advance the paradigm of antitumor immunity in colorectal cancer and provide a blueprint for the detailed characterization of the involved immune cell subsets. The coordinated action of innate and adaptive immune cell populations suggests a multitargeted exploitation of their antitumor properties in a therapeutic setting. Citation Format: Noel F. de Miranda, Natasja L. de Vries, Vincent van Unen, Tamim Abdelaal, Marieke E. Ijsselsteijn, Ruud van der Breggen, Arantza Farina-Sarasqueta, Koen C.M.J. Peeters, Thomas Hollt, Boudewijn P.F. Lelieveldt, Frits Koning. Multidimensional cytometric analysis of colorectal cancer reveals novel and diverse mediators of antitumor immunity [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translat","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"88073481","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A055
M. Casanova
Macrophages are the professional phagocytes of the innate immune system required to remove cellular debris and maintain tissue homeostasis. However, they can also play a role in the pathophysiology of many diseases. In non-small-cell lung carcinoma (NSCLC), macrophages constitute one of the major leukocyte subsets founds in tumors and contribute to immunosuppression by releasing growth factors, inhibiting immune T-cell surveillance, and enhancing angiogenesis, among other mechanisms. Importantly, macrophages in the lung can arise from embryonic or from adult hematopoietic precursors derived from the bone marrow (BM). This dual source of phagocytes has long been ignored and their contribution to tumor progression and response to immunotherapy has remained largely unexplored. Here we propose to analyze the role of macrophages during early and late stages of NSCLC. Using lineage-tracing murine models, tumor spheroids, and high-dimensional techniques to capture macrophage heterogeneity within the lesions (scRNAseq), we found that embryonic macrophages are progressively excluded from the tumor mass, whereas BM-derived ones are evenly distributed. Depletion of embryonic macrophages in a pretumor implantation setting diminishes tumor size and metastasis and leads to an increase in T-cell infiltration and antitumor activity, whereas elimination of tissue-resident macrophages post-tumor implantation does not impact tumor burden or immunity. Importantly, early human NSCLC lesions (stage I) show reduced macrophage infiltrates within the tumor microenvironment, which correlate with poor T-cell infiltration. Our findings suggest that macrophage ontogeny plays a pivotal role in tumor immunity. In this context, we aim to develop novel immunotherapies that harness embryonic macrophages to prevent tumor progression during early phases of lung cancer. Citation Format: Maria Casanova. Lineage–tracing reveals a unique contribution of embryonic macrophages to NSCLC progression [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A055.
{"title":"Abstract A055: Lineage–tracing reveals a unique contribution of embryonic macrophages to NSCLC progression","authors":"M. Casanova","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A055","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A055","url":null,"abstract":"Macrophages are the professional phagocytes of the innate immune system required to remove cellular debris and maintain tissue homeostasis. However, they can also play a role in the pathophysiology of many diseases. In non-small-cell lung carcinoma (NSCLC), macrophages constitute one of the major leukocyte subsets founds in tumors and contribute to immunosuppression by releasing growth factors, inhibiting immune T-cell surveillance, and enhancing angiogenesis, among other mechanisms. Importantly, macrophages in the lung can arise from embryonic or from adult hematopoietic precursors derived from the bone marrow (BM). This dual source of phagocytes has long been ignored and their contribution to tumor progression and response to immunotherapy has remained largely unexplored. Here we propose to analyze the role of macrophages during early and late stages of NSCLC. Using lineage-tracing murine models, tumor spheroids, and high-dimensional techniques to capture macrophage heterogeneity within the lesions (scRNAseq), we found that embryonic macrophages are progressively excluded from the tumor mass, whereas BM-derived ones are evenly distributed. Depletion of embryonic macrophages in a pretumor implantation setting diminishes tumor size and metastasis and leads to an increase in T-cell infiltration and antitumor activity, whereas elimination of tissue-resident macrophages post-tumor implantation does not impact tumor burden or immunity. Importantly, early human NSCLC lesions (stage I) show reduced macrophage infiltrates within the tumor microenvironment, which correlate with poor T-cell infiltration. Our findings suggest that macrophage ontogeny plays a pivotal role in tumor immunity. In this context, we aim to develop novel immunotherapies that harness embryonic macrophages to prevent tumor progression during early phases of lung cancer. Citation Format: Maria Casanova. Lineage–tracing reveals a unique contribution of embryonic macrophages to NSCLC progression [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A055.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"91073134","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A115
Nilesh P. Talele, Hao Liu, K. Naxerova, M. Pinter, J. Incio, Hang Lee, Kohei Shigeta, William W. Ho, T. Michelakos, T. Hong, Jeffrey W. Clark, Janet E. Murphy, D. Ryan, V. Deshpande, K. Lillemoe, C. F. Castillo, M. Downes, R. Evans, J. Michaelson, D. Duda, C. Ferrone, Yves Bouches, R. Jain
Purpose: Angiotensin system inhibitors (ASIs) can improve prognosis in multiple cancer types, including pancreatic ductal adenocarcinoma (PDAC). However, no study has examined the effect of ASIs alone or combined with adjuvant chemotherapy in resected PDAC patients. Experimental Design: We performed an analysis of the records of ASI users and non-user patients with PDAC seen at Massachusetts General Hospital between January 2006 and December 2010. To identify mechanisms of ASIs in PDAC, we performed RNA-Seq of resected primary lesions. In a prospective trial (NCT01821729), we investigated circulating biomarkers of losartan activity. Results: 794 consecutive patients were included. In 299 resected patients, ASI-users experienced longer overall survival (OS) in both univariate (median OS: 36.3 vs. 19.3 months, p=0.011) and adjusted multivariate (HR, 0.505; 95%CI, 0.339 - 0.750; p=0.001) analyses. Propensity score adjusted analysis also showed a longer median OS for chronic ASI users. In unresected patients, the beneficial effect of ASIs was significant in patients with locally advanced disease, but not in metastatic patients. RNA-Seq analysis revealed in tumors of ASI-users (lisinopril) a normalized extracellular matrix, a reduced expression of genes involved in PDAC progression (e.g., WNT and Notch signaling) and an increased expression of genes linked with the activity of T-cells and antigen-presenting cells. Finally, chronic use of ASI was associated with a gene expression signature that is predictive of survival in independent validation cohorts. Circulating biomarker data will be presented at the meeting. Conclusions: In patients with non-metastatic PDAC, chronic ASI use is associated with longer OS independently of chemotherapy. Our RNA-Seq analysis suggests that ASI reduce the malignant potential of cancer cells and stimulate the immune microenvironment in primary PDAC. Citation Format: Nilesh P. Talele, Hao Liu, Kamila Naxerova, Matthias Pinter, Joao Incio, Hang Lee, Kohei Shigeta, William W. Ho, Theodoros Michelakos, Theodore S. Hong, Jeffrey W. Clark, Janet E. Murphy, David P. Ryan, Vikram Deshpande, Kieth D. Lillemoe, Carlos Fernandez-del Castillo, Michael Downes, Ronald M. Evans, James Michaelson, Dan G. Duda, Cristina R. Ferrone, Yves Bouches, Rakesh Jain. Use of angiotensin system inhibitors is associated with immune activation and longer survival in pancreatic ductal adenocarcinoma patients [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A115.
目的:血管紧张素系统抑制剂(ASIs)可以改善多种癌症类型的预后,包括胰腺导管腺癌(PDAC)。然而,目前还没有研究考察ASIs单独或联合辅助化疗对PDAC切除患者的影响。实验设计:我们对2006年1月至2010年12月在马萨诸塞州总医院(Massachusetts General Hospital)就诊的ASI使用者和非ASI使用者PDAC患者的记录进行了分析。为了确定ASIs在PDAC中的机制,我们对切除的原发病变进行了rna测序。在一项前瞻性试验(NCT01821729)中,我们调查了氯沙坦活性的循环生物标志物。结果:连续纳入794例患者。在299例手术切除的患者中,单因素(中位生存期:36.3个月vs. 19.3个月,p=0.011)和调整多因素(HR, 0.505;95%ci, 0.339 - 0.750;p = 0.001)分析。倾向评分调整分析也显示慢性ASI使用者的中位OS更长。在未切除的患者中,ASIs的有益效果在局部晚期患者中显着,而在转移患者中则不明显。RNA-Seq分析显示,在asi使用者(赖诺普利)的肿瘤中,细胞外基质正常化,参与PDAC进展的基因表达减少(例如,WNT和Notch信号),与t细胞和抗原呈递细胞活性相关的基因表达增加。最后,在独立验证队列中,长期使用ASI与预测生存的基因表达特征相关。将在会议上介绍流行的生物标志物数据。结论:在非转移性PDAC患者中,慢性ASI使用与化疗无关的更长的生存期相关。我们的RNA-Seq分析表明,ASI降低了癌细胞的恶性潜能,并刺激了原发性PDAC的免疫微环境。引文格式:Nilesh P. Talele, Hao Liu, Kamila Naxerova, Matthias Pinter, Joao Incio, Hang Lee, Kohei Shigeta, William W. Ho, Theodoros Michelakos, Theodore S. Hong, Jeffrey W. Clark, Janet E. Murphy, David P. Ryan, Vikram Deshpande, Kieth D. Lillemoe, Carlos Fernandez-del Castillo, Michael Downes, Ronald M. Evans, James Michaelson, Dan G. Duda, Cristina R. Ferrone, Yves Bouches, Rakesh Jain。血管紧张素系统抑制剂的使用与胰腺导管腺癌患者的免疫激活和更长的生存期有关[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫,2019;7(2增刊):摘要nr A115。
{"title":"Abstract A115: Use of angiotensin system inhibitors is associated with immune activation and longer survival in pancreatic ductal adenocarcinoma patients","authors":"Nilesh P. Talele, Hao Liu, K. Naxerova, M. Pinter, J. Incio, Hang Lee, Kohei Shigeta, William W. Ho, T. Michelakos, T. Hong, Jeffrey W. Clark, Janet E. Murphy, D. Ryan, V. Deshpande, K. Lillemoe, C. F. Castillo, M. Downes, R. Evans, J. Michaelson, D. Duda, C. Ferrone, Yves Bouches, R. Jain","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A115","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A115","url":null,"abstract":"Purpose: Angiotensin system inhibitors (ASIs) can improve prognosis in multiple cancer types, including pancreatic ductal adenocarcinoma (PDAC). However, no study has examined the effect of ASIs alone or combined with adjuvant chemotherapy in resected PDAC patients. Experimental Design: We performed an analysis of the records of ASI users and non-user patients with PDAC seen at Massachusetts General Hospital between January 2006 and December 2010. To identify mechanisms of ASIs in PDAC, we performed RNA-Seq of resected primary lesions. In a prospective trial (NCT01821729), we investigated circulating biomarkers of losartan activity. Results: 794 consecutive patients were included. In 299 resected patients, ASI-users experienced longer overall survival (OS) in both univariate (median OS: 36.3 vs. 19.3 months, p=0.011) and adjusted multivariate (HR, 0.505; 95%CI, 0.339 - 0.750; p=0.001) analyses. Propensity score adjusted analysis also showed a longer median OS for chronic ASI users. In unresected patients, the beneficial effect of ASIs was significant in patients with locally advanced disease, but not in metastatic patients. RNA-Seq analysis revealed in tumors of ASI-users (lisinopril) a normalized extracellular matrix, a reduced expression of genes involved in PDAC progression (e.g., WNT and Notch signaling) and an increased expression of genes linked with the activity of T-cells and antigen-presenting cells. Finally, chronic use of ASI was associated with a gene expression signature that is predictive of survival in independent validation cohorts. Circulating biomarker data will be presented at the meeting. Conclusions: In patients with non-metastatic PDAC, chronic ASI use is associated with longer OS independently of chemotherapy. Our RNA-Seq analysis suggests that ASI reduce the malignant potential of cancer cells and stimulate the immune microenvironment in primary PDAC. Citation Format: Nilesh P. Talele, Hao Liu, Kamila Naxerova, Matthias Pinter, Joao Incio, Hang Lee, Kohei Shigeta, William W. Ho, Theodoros Michelakos, Theodore S. Hong, Jeffrey W. Clark, Janet E. Murphy, David P. Ryan, Vikram Deshpande, Kieth D. Lillemoe, Carlos Fernandez-del Castillo, Michael Downes, Ronald M. Evans, James Michaelson, Dan G. Duda, Cristina R. Ferrone, Yves Bouches, Rakesh Jain. Use of angiotensin system inhibitors is associated with immune activation and longer survival in pancreatic ductal adenocarcinoma patients [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A115.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83533993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A113
K. Silina, A. Soltermann, Chiara Burkhardt, Farkhondeh Movahedian Attar, R. Casanova, A. Curioni-Fontecedro, H. Moch, F. Posch, T. Winder, N. V. Dijk, C. Voskuilen, M. S. Heijden, M. Broek
Lymphoid organ neogenesis takes place in chronically inflamed tissues including cancer and yields the development of tertiary lymphoid structures (TLS). TLS are ectopic lymphoid organs that activate antigen specific T-cells and B cells in infection and autoimmunity and correlate with prolonged survival in various cancer types. This suggests that TLS contribute to protective anti-tumor immunity. Therefore, promoting the development of tumor-associated TLS could be a novel immunotherapeutic approach. However, the molecular and cellular mechanisms of TLS development in human cancer or how TLS contribute to survival are largely not understood. Here we used multiparameter immunofluorescence and digital pathology to quantify TLS and to characterize their cellular composition and tissue context in cohorts of lung squamous cell carcinoma (LSCC, n=138), colorectal cancer (CRC, n=111), clear cell renal cell carcinoma (ccRCC, n=50) and bladder cancer (BC, n=33) patients. Furthermore, we established an experimental model to characterize TLS development and its impact on tumor-specific immunity. We discovered that TLS development and maturation followed the same steps in all analyzed tumor types as well as in the lungs of mice in our experimental model. First, B and T lymphocytes accumulated around blood vessels. Second, a network of follicular dendritic cells developed within the lymphocytic aggregate, and third, a germinal center (GC) reaction was activated. Additionally, we identified a niche of CXCL13+ perivascular stroma and CXCL12+LTB+ and PD-L1+ epithelial cells that were associated with TLS in LSCC. We found that the number of tumor-associated TLS was an independent prognostic factor for prolonged survival in untreated LSCC, CRC and BC, but not in ccRCC patients or in LSCC and BC patients who were treated with neoadjuvant chemotherapy. By comparing the chemotherapy-treated and untreated cohorts we observed that the number of TLS was not changed but TLS maturation (i.e. GC formation) was significantly impaired after chemotherapy. This difference was at least partially dictated by corticosteroids, which are commonly used to treat the side effects of chemotherapy of LSCC patients. We further studied the mechanisms underlying TLS development using the experimental model. We identified a combination of stimuli that induces the development of mature TLS in the lungs of mice. Besides inflammatory stimuli, a foreign antigen was necessary to achieve a significant increase in TLS numbers and maturation stage, suggesting that cognate interactions are crucial for lymphoid organ neogenesis. This is further supported by our observation that CRC patients with microsatellite instability, which presumably results in more neoantigens, had an increased proportion of mature TLS. The negative impact of corticosteroids on TLS development was confirmed in this model. In summary, we propose that GC+ TLS represent the relevant TLS phenotype contributing to survival in differe
淋巴器官新生发生在包括癌症在内的慢性炎症组织中,并产生三级淋巴样结构(TLS)。TLS是异位淋巴器官,在感染和自身免疫中激活抗原特异性t细胞和B细胞,并与各种癌症类型的延长生存相关。这表明TLS参与了保护性抗肿瘤免疫。因此,促进肿瘤相关TLS的发展可能是一种新的免疫治疗方法。然而,TLS在人类癌症中发展的分子和细胞机制以及TLS如何促进生存在很大程度上尚不清楚。在这里,我们使用多参数免疫荧光和数字病理学来量化TLS,并表征它们在肺鳞状细胞癌(LSCC, n=138)、结直肠癌(CRC, n=111)、透明细胞肾细胞癌(ccRCC, n=50)和膀胱癌(BC, n=33)患者队列中的细胞组成和组织背景。此外,我们建立了一个实验模型来表征TLS的发展及其对肿瘤特异性免疫的影响。我们发现,在我们的实验模型中,所有分析的肿瘤类型以及小鼠的肺部中,TLS的发育和成熟遵循相同的步骤。首先,B淋巴细胞和T淋巴细胞聚集在血管周围。第二,滤泡树突状细胞网络在淋巴细胞聚集体内发育,第三,生发中心(GC)反应被激活。此外,我们还发现了与LSCC中TLS相关的CXCL13+血管周围基质和CXCL12+LTB+和PD-L1+上皮细胞。我们发现肿瘤相关TLS的数量是未经治疗的LSCC、CRC和BC患者延长生存期的独立预后因素,但在ccRCC患者或接受新辅助化疗的LSCC和BC患者中则不是。通过比较化疗组和未化疗组,我们发现化疗后TLS的数量没有改变,但TLS的成熟(即GC的形成)明显受损。这种差异至少部分是由皮质类固醇决定的,皮质类固醇通常用于治疗LSCC患者化疗的副作用。我们使用实验模型进一步研究了TLS开发的机制。我们确定了一种刺激组合,可诱导小鼠肺部成熟TLS的发育。除了炎症刺激外,外源抗原是实现TLS数量和成熟阶段显著增加所必需的,这表明同源相互作用对淋巴器官新生至关重要。我们的观察结果进一步支持了这一点,即微卫星不稳定的CRC患者成熟TLS的比例增加,这可能导致更多的新抗原。该模型证实了皮质类固醇对TLS发育的负面影响。综上所述,我们认为GC+ TLS代表了在不同肿瘤类型中对生存有贡献的相关TLS表型。皮质类固醇对淋巴器官新生有负面影响,可能损害自发和治疗诱导的抗肿瘤免疫。建立的实验模型将允许研究TLS在癌症中的发育和功能机制,并评估其治疗潜力。引文格式:Karina Silina, Alex Soltermann, Chiara Burkhardt, Farkhondeh Movahedian Attar, Ruben Casanova, Alessandra Curioni-Fontecedro, Holger Moch, Florian Posch, Thomas Winder, Nick van Dijk, Charlotte Voskuilen, michel van der Heijden, Maries van den Broek。利用淋巴器官新生作为新的预后生物标志物和治疗靶点[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫,2019;7(2增刊):摘要nr A113。
{"title":"Abstract A113: Harnessing lymphoid organ neogenesis as a novel prognostic biomarker and therapeutic target","authors":"K. Silina, A. Soltermann, Chiara Burkhardt, Farkhondeh Movahedian Attar, R. Casanova, A. Curioni-Fontecedro, H. Moch, F. Posch, T. Winder, N. V. Dijk, C. Voskuilen, M. S. Heijden, M. Broek","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A113","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A113","url":null,"abstract":"Lymphoid organ neogenesis takes place in chronically inflamed tissues including cancer and yields the development of tertiary lymphoid structures (TLS). TLS are ectopic lymphoid organs that activate antigen specific T-cells and B cells in infection and autoimmunity and correlate with prolonged survival in various cancer types. This suggests that TLS contribute to protective anti-tumor immunity. Therefore, promoting the development of tumor-associated TLS could be a novel immunotherapeutic approach. However, the molecular and cellular mechanisms of TLS development in human cancer or how TLS contribute to survival are largely not understood. Here we used multiparameter immunofluorescence and digital pathology to quantify TLS and to characterize their cellular composition and tissue context in cohorts of lung squamous cell carcinoma (LSCC, n=138), colorectal cancer (CRC, n=111), clear cell renal cell carcinoma (ccRCC, n=50) and bladder cancer (BC, n=33) patients. Furthermore, we established an experimental model to characterize TLS development and its impact on tumor-specific immunity. We discovered that TLS development and maturation followed the same steps in all analyzed tumor types as well as in the lungs of mice in our experimental model. First, B and T lymphocytes accumulated around blood vessels. Second, a network of follicular dendritic cells developed within the lymphocytic aggregate, and third, a germinal center (GC) reaction was activated. Additionally, we identified a niche of CXCL13+ perivascular stroma and CXCL12+LTB+ and PD-L1+ epithelial cells that were associated with TLS in LSCC. We found that the number of tumor-associated TLS was an independent prognostic factor for prolonged survival in untreated LSCC, CRC and BC, but not in ccRCC patients or in LSCC and BC patients who were treated with neoadjuvant chemotherapy. By comparing the chemotherapy-treated and untreated cohorts we observed that the number of TLS was not changed but TLS maturation (i.e. GC formation) was significantly impaired after chemotherapy. This difference was at least partially dictated by corticosteroids, which are commonly used to treat the side effects of chemotherapy of LSCC patients. We further studied the mechanisms underlying TLS development using the experimental model. We identified a combination of stimuli that induces the development of mature TLS in the lungs of mice. Besides inflammatory stimuli, a foreign antigen was necessary to achieve a significant increase in TLS numbers and maturation stage, suggesting that cognate interactions are crucial for lymphoid organ neogenesis. This is further supported by our observation that CRC patients with microsatellite instability, which presumably results in more neoantigens, had an increased proportion of mature TLS. The negative impact of corticosteroids on TLS development was confirmed in this model. In summary, we propose that GC+ TLS represent the relevant TLS phenotype contributing to survival in differe","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72650445","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}