首页 > 最新文献

Tackling the Tumor Microenvironment: Beyond T-cells最新文献

英文 中文
Abstract A110: Efficacy of anti-PD1 immune checkpoint blockade involves the cooperative interaction of myeloid and lymphoid subpopulations in the tumor microenvironment A110:抗pd1免疫检查点阻断的有效性涉及肿瘤微环境中髓系和淋巴系亚群的协同相互作用
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A110
Sjoerd T. T. Schetters, Y. Kooyk
Suppression of the immune system by solid malignancies has proven to be a driving force of tumor development and an effective target for therapeutic intervention. The suppression of cytolytic T-cells by inhibitory receptors like PD1 can be blocked by antagonistic antibodies, reinvigorating suppressed antitumor responses. Nonetheless, only a minority of patients show clinical benefit. It is becoming clear the efficacy of checkpoint blockade relies on many factors, including pretreatment conditions, collaboration between innate and adaptive immune cells and immune-architecture of the tumor microenvironment (TME). To investigate this, we studied the immune profiles of the PD1-unresponsive murine B16 melanoma and PD1-responsive MC38 colorectal carcinoma models, systemically and in the TME before and during treatment. By using high-dimensional flow cytometry and unsupervised clustering analyses based on immune checkpoints, we show comparable early establishment of heterogeneity of tumor-infiltrating CD8+ and CD4+ T-cells. However, PD1-responsive MC38 tumor shows correlations in abundance between specific CD8+ T-cell, NK cells and myeloid subsets before checkpoint blockade treatment, while the PD1-unresponsive B16 tumors do not show lymphoid-myeloid codependences. Interestingly, the abundance of monocyte-derived dendritic cells did not increase upon anti-PD1 treatment but instead showed abundance correlation with PD1 CD4+ and CD8+ T-cells, suggesting a putative interaction. The unresponsive B16 tumors showed increased correlation of cDC1 and cDC2 with Foxp3+ regulatory T-cells. We have visualized these putative interactions within the myeloid and lymphoid population in the changing immune-architecture of the TME, using multiplex fluorescence and confocal microscopy. We show heterogeneity and interactive hotspots during immune checkpoint blockade and postulate that successful anti-PD1 treatment requires the location-dependent cooperation of specific myeloid and lymphoid subsets in the TME. Citation Format: Sjoerd Schetters, Yvette Van Kooyk. Efficacy of anti-PD1 immune checkpoint blockade involves the cooperative interaction of myeloid and lymphoid subpopulations in the 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 A110.
实体恶性肿瘤对免疫系统的抑制已被证明是肿瘤发展的驱动力和治疗干预的有效靶点。抑制受体如PD1对细胞溶解性t细胞的抑制可以被拮抗抗体阻断,从而使抑制的抗肿瘤反应重新活跃起来。然而,只有少数患者表现出临床获益。检查点阻断的有效性取决于许多因素,包括预处理条件、先天和适应性免疫细胞之间的协作以及肿瘤微环境(TME)的免疫结构,这一点越来越清楚。为了研究这一点,我们研究了pd1无应答的小鼠B16黑色素瘤和pd1应答的MC38结直肠癌模型在治疗前和治疗期间的全身和TME的免疫谱。通过使用高维流式细胞术和基于免疫检查点的无监督聚类分析,我们显示了肿瘤浸润性CD8+和CD4+ t细胞的早期异质性。然而,在检查点阻断治疗前,pd1应答的MC38肿瘤显示特异性CD8+ t细胞、NK细胞和髓细胞亚群之间存在丰富的相关性,而pd1无应答的B16肿瘤不显示淋巴-髓细胞共依赖性。有趣的是,单核细胞来源的树突状细胞的丰度在抗PD1治疗后并没有增加,而是与PD1 CD4+和CD8+ t细胞的丰度相关,这表明可能存在相互作用。无应答的B16肿瘤显示cDC1和cDC2与Foxp3+调节性t细胞的相关性增加。我们利用多重荧光和共聚焦显微镜观察了骨髓和淋巴细胞群体在TME免疫结构变化中的这些假定的相互作用。我们在免疫检查点阻断过程中发现了异质性和相互作用热点,并假设成功的抗pd1治疗需要TME中特定髓细胞和淋巴细胞亚群的位置依赖性合作。引文格式:Sjoerd Schetters, Yvette Van Kooyk。抗pd1免疫检查点阻断的有效性涉及肿瘤微环境中髓系和淋巴系亚群的协同相互作用[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 A110。
{"title":"Abstract A110: Efficacy of anti-PD1 immune checkpoint blockade involves the cooperative interaction of myeloid and lymphoid subpopulations in the tumor microenvironment","authors":"Sjoerd T. T. Schetters, Y. Kooyk","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A110","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A110","url":null,"abstract":"Suppression of the immune system by solid malignancies has proven to be a driving force of tumor development and an effective target for therapeutic intervention. The suppression of cytolytic T-cells by inhibitory receptors like PD1 can be blocked by antagonistic antibodies, reinvigorating suppressed antitumor responses. Nonetheless, only a minority of patients show clinical benefit. It is becoming clear the efficacy of checkpoint blockade relies on many factors, including pretreatment conditions, collaboration between innate and adaptive immune cells and immune-architecture of the tumor microenvironment (TME). To investigate this, we studied the immune profiles of the PD1-unresponsive murine B16 melanoma and PD1-responsive MC38 colorectal carcinoma models, systemically and in the TME before and during treatment. By using high-dimensional flow cytometry and unsupervised clustering analyses based on immune checkpoints, we show comparable early establishment of heterogeneity of tumor-infiltrating CD8+ and CD4+ T-cells. However, PD1-responsive MC38 tumor shows correlations in abundance between specific CD8+ T-cell, NK cells and myeloid subsets before checkpoint blockade treatment, while the PD1-unresponsive B16 tumors do not show lymphoid-myeloid codependences. Interestingly, the abundance of monocyte-derived dendritic cells did not increase upon anti-PD1 treatment but instead showed abundance correlation with PD1 CD4+ and CD8+ T-cells, suggesting a putative interaction. The unresponsive B16 tumors showed increased correlation of cDC1 and cDC2 with Foxp3+ regulatory T-cells. We have visualized these putative interactions within the myeloid and lymphoid population in the changing immune-architecture of the TME, using multiplex fluorescence and confocal microscopy. We show heterogeneity and interactive hotspots during immune checkpoint blockade and postulate that successful anti-PD1 treatment requires the location-dependent cooperation of specific myeloid and lymphoid subsets in the TME. Citation Format: Sjoerd Schetters, Yvette Van Kooyk. Efficacy of anti-PD1 immune checkpoint blockade involves the cooperative interaction of myeloid and lymphoid subpopulations in the 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 A110.","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":"90303561","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}
引用次数: 0
Abstract A082: Single-cell RNA-sequencing of metastatic melanoma identifies a cancer cell-intrinsic program associated with immune checkpoint inhibitor resistance 摘要:转移性黑色素瘤的单细胞rna测序鉴定出与免疫检查点抑制剂耐药性相关的癌细胞内在程序
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A082
Livnat Jerby, P. Shah, Michael S. Cuoco, Christopher Rodman, Mei-ju Su, Johannes Melms, Rachel Leeson, Abhay Kanodia, Shaolin Mei, Jia-Ren Lin, Shu Wang, Bokang Rabasha, David Liu, Gao Zhang, Claire Margolais, Orr Ashenberg, P. Ott, E. Buchbinder, R. Haq, Stephen F. Hodi, G. Boland, R. Sullivan, Dennie T. Frederick, B. Miao, Tabea Moll, K. Flaherty, M. Herlyn, R. Jenkins, Rohit Thummalapalli, Monika S. Kowalczyk, I. Canadas, B. Schilling, Adam N. Cartwright, Adrienne M. Luoma, Shruti Malu, P. Hwu, C. Bernatchez, M. Forget, D. Barbie, A. Shalek, I. Tirosh, P. Sorger, K. Wucherpfennig, E. Allen, D. Schadendorf, B. Johnson, Asaf Rotem, Orit Rosenblatt-Rozen, L. Garraway, Charles H. Yoon, B. Izar, A. Regev
Immune checkpoint inhibitors (ICI) produce durable responses in some melanoma patients, but many patients derive no clinical benefit. The molecular underpinnings of ICI resistance involve intricate cell-cell interactions that are yet elusive. To systematically map the interactions between malignant and immune cells in the tumor ecosystem, we applied single-cell RNA sequencing to 31 human melanoma tumors, profiling thousands of malignant, immune, and stromal cells. We identified a transcriptional program in malignanT-cells that is strongly associated with T-cell exclusion and immunotherapy resistance. Using highly multiplexed in situ imaging we first demonstrated that this program characterizes malignanT-cells in “cold” niches. Next, we showed that the program predicts clinical responses to ICI according to multiple independent validation cohorts, including a new cohort that we obtained from 112 melanoma patients treated with anti-PD-1 therapy. We then identified CDK4/6 as master regulators of this resistance program, and found that CDK4/6 inhibitors repress the program and shift melanoma cells into a senescence-associated secretory phenotype. Lastly, we showed that CDK4/6-inhibition leads to a substantial reduction in melanoma tumor outgrowth in a B16 mouse model when given in combination with immunotherapy. Taken together, our study provides a high-resolution landscape of ICI-resistant cell states, identifies clinically predictive signatures, and forms a basis for the development of novel therapeutic strategies that could overcome immunotherapy resistance. Citation Format: Livnat Jerby, Parin Shah, Michael S. Cuoco, Christopher Rodman, Mei-Ju Su, Johannes M. Melms, Rachel Leeson, Abhay Kanodia, Shaolin Mei, Jia-Ren Lin, Shu Wang, Bokang Rabasha, David Liu, Gao Zhang, Claire Margolais, Orr Ashenberg, Patrick A. Ott, Elizabeth I. Buchbinder, Riz Haq, Stephen Hodi, Genevieve M. Boland, Ryan J. Sullivan, Dennie Frederick, Benchun Miao, Tabea Moll, Keith Flaherty, Meenhard Herlyn, Russell S. Jenkins, Rohit Thummalapalli, Monika S. Kowalczyk, Israel Canadas, Bastian Schilling, Adam N.R Cartwright, Adrienne M. Luoma, Shruti Malu, Patrick Hwu, Chantale Bernatchez, Marie-Andree Forget, David A. Barbie, Alex K. Shalek, Itay Tirosh, Peter K. Sorger, Kai W. Wucherpfennig, Eliezer M. Van Allen, Dirk Schadendorf, Bruce E. Johnson, Asaf Rotem, Orit Rosenblatt-Rozen, Levi A. Garraway, Charles H. Yoon, Benjamin Izar, Aviv Regev. Single-cell RNA-sequencing of metastatic melanoma identifies a cancer cell-intrinsic program associated with immune checkpoint inhibitor resistance [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 A082.
免疫检查点抑制剂(ICI)在一些黑色素瘤患者中产生持久的反应,但许多患者没有获得临床益处。ICI抗性的分子基础涉及复杂的细胞-细胞相互作用,但仍难以捉摸。为了系统地绘制肿瘤生态系统中恶性细胞和免疫细胞之间的相互作用,我们对31个人类黑色素瘤肿瘤应用单细胞RNA测序,分析了数千个恶性、免疫和基质细胞。我们在恶性抗细胞中发现了一个转录程序,它与t细胞排斥和免疫治疗耐药性密切相关。使用高复用原位成像,我们首先证明了该程序表征了“冷”壁龛中的恶性抗细胞。接下来,我们展示了该程序根据多个独立验证队列预测对ICI的临床反应,包括我们从112名接受抗pd -1治疗的黑色素瘤患者中获得的新队列。然后,我们确定CDK4/6是该耐药程序的主要调节因子,并发现CDK4/6抑制剂抑制该程序并将黑色素瘤细胞转变为衰老相关的分泌表型。最后,我们发现在B16小鼠模型中,cdk4 /6抑制与免疫治疗联合使用可显著减少黑色素瘤的生长。总之,我们的研究提供了ici耐药细胞状态的高分辨率景观,确定了临床预测特征,并为开发可以克服免疫治疗耐药的新治疗策略奠定了基础。引文格式:Livnat Jerby, Parin Shah, Michael S. Cuoco, Christopher Rodman, Su Mei- ju, Johannes M. Melms, Rachel Leeson, Abhay Kanodia, me少林,林家仁,王舒,Bokang Rabasha, David Liu, Zhang Gao, Claire Margolais, Orr Ashenberg, Patrick A. Ott, Elizabeth I. Buchbinder, Riz Haq, Stephen Hodi, Genevieve M. Boland, Ryan J. Sullivan, Dennie Frederick, Benchun Miao, Tabea Moll, Keith Flaherty, Meenhard Herlyn, Russell S. Jenkins, Rohit Thummalapalli, Monika S. Kowalczyk, Israel加拿大,Bastian Schilling, Adam N.R Cartwright, Adrienne M. Luoma, Shruti Malu, Patrick Hwu, Chantale Bernatchez, Marie-Andree Forget, David A. Barbie, Alex K. Shalek, Itay Tirosh, Peter K. Sorger, Kai W. Wucherpfennig, Eliezer M. Van Allen, Dirk Schadendorf, Bruce E. Johnson, Asaf Rotem, Orit rosenblat - rozen, Levi A. Garraway, Charles H. Yoon, Benjamin Izar, Aviv Regev。转移性黑色素瘤的单细胞rna测序鉴定出与免疫检查点抑制剂耐药性相关的癌细胞内在程序[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 A082。
{"title":"Abstract A082: Single-cell RNA-sequencing of metastatic melanoma identifies a cancer cell-intrinsic program associated with immune checkpoint inhibitor resistance","authors":"Livnat Jerby, P. Shah, Michael S. Cuoco, Christopher Rodman, Mei-ju Su, Johannes Melms, Rachel Leeson, Abhay Kanodia, Shaolin Mei, Jia-Ren Lin, Shu Wang, Bokang Rabasha, David Liu, Gao Zhang, Claire Margolais, Orr Ashenberg, P. Ott, E. Buchbinder, R. Haq, Stephen F. Hodi, G. Boland, R. Sullivan, Dennie T. Frederick, B. Miao, Tabea Moll, K. Flaherty, M. Herlyn, R. Jenkins, Rohit Thummalapalli, Monika S. Kowalczyk, I. Canadas, B. Schilling, Adam N. Cartwright, Adrienne M. Luoma, Shruti Malu, P. Hwu, C. Bernatchez, M. Forget, D. Barbie, A. Shalek, I. Tirosh, P. Sorger, K. Wucherpfennig, E. Allen, D. Schadendorf, B. Johnson, Asaf Rotem, Orit Rosenblatt-Rozen, L. Garraway, Charles H. Yoon, B. Izar, A. Regev","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A082","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A082","url":null,"abstract":"Immune checkpoint inhibitors (ICI) produce durable responses in some melanoma patients, but many patients derive no clinical benefit. The molecular underpinnings of ICI resistance involve intricate cell-cell interactions that are yet elusive. To systematically map the interactions between malignant and immune cells in the tumor ecosystem, we applied single-cell RNA sequencing to 31 human melanoma tumors, profiling thousands of malignant, immune, and stromal cells. We identified a transcriptional program in malignanT-cells that is strongly associated with T-cell exclusion and immunotherapy resistance. Using highly multiplexed in situ imaging we first demonstrated that this program characterizes malignanT-cells in “cold” niches. Next, we showed that the program predicts clinical responses to ICI according to multiple independent validation cohorts, including a new cohort that we obtained from 112 melanoma patients treated with anti-PD-1 therapy. We then identified CDK4/6 as master regulators of this resistance program, and found that CDK4/6 inhibitors repress the program and shift melanoma cells into a senescence-associated secretory phenotype. Lastly, we showed that CDK4/6-inhibition leads to a substantial reduction in melanoma tumor outgrowth in a B16 mouse model when given in combination with immunotherapy. Taken together, our study provides a high-resolution landscape of ICI-resistant cell states, identifies clinically predictive signatures, and forms a basis for the development of novel therapeutic strategies that could overcome immunotherapy resistance. Citation Format: Livnat Jerby, Parin Shah, Michael S. Cuoco, Christopher Rodman, Mei-Ju Su, Johannes M. Melms, Rachel Leeson, Abhay Kanodia, Shaolin Mei, Jia-Ren Lin, Shu Wang, Bokang Rabasha, David Liu, Gao Zhang, Claire Margolais, Orr Ashenberg, Patrick A. Ott, Elizabeth I. Buchbinder, Riz Haq, Stephen Hodi, Genevieve M. Boland, Ryan J. Sullivan, Dennie Frederick, Benchun Miao, Tabea Moll, Keith Flaherty, Meenhard Herlyn, Russell S. Jenkins, Rohit Thummalapalli, Monika S. Kowalczyk, Israel Canadas, Bastian Schilling, Adam N.R Cartwright, Adrienne M. Luoma, Shruti Malu, Patrick Hwu, Chantale Bernatchez, Marie-Andree Forget, David A. Barbie, Alex K. Shalek, Itay Tirosh, Peter K. Sorger, Kai W. Wucherpfennig, Eliezer M. Van Allen, Dirk Schadendorf, Bruce E. Johnson, Asaf Rotem, Orit Rosenblatt-Rozen, Levi A. Garraway, Charles H. Yoon, Benjamin Izar, Aviv Regev. Single-cell RNA-sequencing of metastatic melanoma identifies a cancer cell-intrinsic program associated with immune checkpoint inhibitor resistance [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 A082.","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":"85331088","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}
引用次数: 0
Abstract A086: Omental fat in ovarian cancer induces lymphangiogenesis A086:卵巢癌大网膜脂肪诱导淋巴管生成
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A086
B. Lenoir, D. Ferber, V. Starrach, M. Suarez-Carmona, S. Schott, I. Zoernig, D. Jäger, N. Halama
Ovarian cancer metastasis occurs by direct multifocal seeding into the peritoneum as well as by migration through the lymphatic system. High-grade ovarian carcinoma patients often present with distant metastases. Significant risk factors for the development of those are stage, grade, and lymph node involvement. The increase of the number of lymphatic vessels seems to be implicated in ovarian tumor progression. While the tropism of ovarian cancer cells for fat is well described, the potential impact of an adipose-rich microenvironment on the dissemination of metastasis via lymphatic vessels has never been investigated. So far, in this study, we examined the effect of omental fat on lymphangiogenesis in ovarian carcinoma. For that we used a cohort of 80 ovarian cancer specimens. We observed a higher number of tumor-associated vessels and principally lymphatic vessels in ovarian cancer in contact with the omentum. These lymphatic vessels are predominantly localized along the fat tissue. A higher secretion of VEGF-C is observed in ovarian tissues containing fat compared to the ones without fat, giving a potential explanation to the observed increase of lymphatic vessels in fatty tissues. We also developed a healthy fat tissue explant culture model and treated whole tissue explants with ascites. Herein, we saw an increase of the number of adipose-derived stem cells (ADSCs). These ADSCs express lymphatic markers such as D2-40 and Lyve-1. We also observed an impact of fat supernatant on the proliferation, migration and tube formation of lymphatic endothelial cells in vitro. In conclusion, we can say that omental fat in ovarian cancer seems to have an impact on lymphangiogenesis. The close contact of ascites with fat tissue seems to lead to a differentiation of adipose-derived stem cells into lymphatic endothelial cells. Further investigations must be performed to understand the exact mechanisms underlying this phenomenon. Citation Format: Benedicte M.A. Lenoir, Dyke Ferber, Victor Starrach, Meggy Suarez-Carmona, Sarah Schott, Inka Zoernig, Dirk Jager, Niels Halama. Omental fat in ovarian cancer induces lymphangiogenesis [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 A086.
卵巢癌转移发生直接多灶播种到腹膜以及通过淋巴系统迁移。高级别卵巢癌患者常伴有远处转移。这些疾病发展的重要危险因素是分期、分级和淋巴结受累。淋巴管数量的增加似乎与卵巢肿瘤的进展有关。虽然卵巢癌细胞对脂肪的趋向性得到了很好的描述,但富含脂肪的微环境对通过淋巴管传播转移的潜在影响从未被研究过。到目前为止,在本研究中,我们研究了大网膜脂肪对卵巢癌淋巴管生成的影响。为此,我们使用了一组80个卵巢癌样本。我们观察到卵巢癌中与网膜接触的肿瘤相关血管数量较多,主要是淋巴管。这些淋巴管主要沿脂肪组织分布。与不含脂肪的卵巢组织相比,在含脂肪的卵巢组织中观察到更高的VEGF-C分泌,这可能解释了脂肪组织中观察到的淋巴管增加。我们还建立了健康脂肪组织外植体培养模型,并用腹水处理整个组织外植体。在这里,我们看到脂肪来源的干细胞(ADSCs)数量的增加。这些ADSCs表达淋巴标记物,如D2-40和Lyve-1。我们还观察了脂肪上清对体外淋巴内皮细胞增殖、迁移和成管的影响。总之,我们可以说卵巢癌的网膜脂肪似乎对淋巴管生成有影响。腹水与脂肪组织的密切接触似乎导致脂肪来源的干细胞分化为淋巴内皮细胞。必须进行进一步的调查,以了解这一现象背后的确切机制。引文格式:Benedicte M.A. Lenoir, Dyke Ferber, Victor Starrach, Meggy Suarez-Carmona, Sarah Schott, Inka zoerning, Dirk Jager, Niels Halama。卵巢癌大网膜脂肪诱导淋巴管生成[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 A086。
{"title":"Abstract A086: Omental fat in ovarian cancer induces lymphangiogenesis","authors":"B. Lenoir, D. Ferber, V. Starrach, M. Suarez-Carmona, S. Schott, I. Zoernig, D. Jäger, N. Halama","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A086","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A086","url":null,"abstract":"Ovarian cancer metastasis occurs by direct multifocal seeding into the peritoneum as well as by migration through the lymphatic system. High-grade ovarian carcinoma patients often present with distant metastases. Significant risk factors for the development of those are stage, grade, and lymph node involvement. The increase of the number of lymphatic vessels seems to be implicated in ovarian tumor progression. While the tropism of ovarian cancer cells for fat is well described, the potential impact of an adipose-rich microenvironment on the dissemination of metastasis via lymphatic vessels has never been investigated. So far, in this study, we examined the effect of omental fat on lymphangiogenesis in ovarian carcinoma. For that we used a cohort of 80 ovarian cancer specimens. We observed a higher number of tumor-associated vessels and principally lymphatic vessels in ovarian cancer in contact with the omentum. These lymphatic vessels are predominantly localized along the fat tissue. A higher secretion of VEGF-C is observed in ovarian tissues containing fat compared to the ones without fat, giving a potential explanation to the observed increase of lymphatic vessels in fatty tissues. We also developed a healthy fat tissue explant culture model and treated whole tissue explants with ascites. Herein, we saw an increase of the number of adipose-derived stem cells (ADSCs). These ADSCs express lymphatic markers such as D2-40 and Lyve-1. We also observed an impact of fat supernatant on the proliferation, migration and tube formation of lymphatic endothelial cells in vitro. In conclusion, we can say that omental fat in ovarian cancer seems to have an impact on lymphangiogenesis. The close contact of ascites with fat tissue seems to lead to a differentiation of adipose-derived stem cells into lymphatic endothelial cells. Further investigations must be performed to understand the exact mechanisms underlying this phenomenon. Citation Format: Benedicte M.A. Lenoir, Dyke Ferber, Victor Starrach, Meggy Suarez-Carmona, Sarah Schott, Inka Zoernig, Dirk Jager, Niels Halama. Omental fat in ovarian cancer induces lymphangiogenesis [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 A086.","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":"76824807","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}
引用次数: 0
Abstract A103: Allosteric inhibition of SHP2 induces antitumor immunity in PD-1-sensitive tumors through modulation of both innate and adaptive mechanisms 摘要:SHP2的变构抑制通过调节先天和适应性机制诱导pd -1敏感肿瘤的抗肿瘤免疫
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A103
E. Quintana, Kasia Mordec, R. Nichols, D. Wildes, C. Schulze, D. Myers, Mallika Singh, E. Koltun, A. Gill, S. Kelsey, M. Goldsmith, Jan Smith
The protein-tyrosine phosphatase SHP2, encoded by PTPN11, is a known oncogenic driver in a subset of cancers and a central signaling node in the RTK-RAS-MAPK pathway. Genetic and pharmacologic evidence supports a role for SHP2 in driving the proliferation of cancer cells dependent upon a range of activated RTKs, certain RAS and BRAF mutations, and NF1 loss of function mutations. In contrast, a role for SHP2 in antitumor immunity is not well established. SHP2 binds to phosphorylated ITIM and ITAM domains on regulatory receptors in immune cells and multiple reports have demonstrated a SHP2/PD-1 physical interaction. Recently it has been proposed that SHP2 transduces the PD-1 inhibitory checkpoint signal by direct de-phosphorylation of CD28. In this study we show that a peptide comprising two tyrosine phosphorylated 9-mers sequences from the PD-1 ITAM (connected by a PEG8 linker) can activate purified SHP2 enzyme. We also demonstrate that, like checkpoint inhibitors, allosteric inhibition of SHP2 activates NFAT-mediated gene expression in a reporter gene PD-1/PD-L1 bioassay. Based on these findings, we evaluated the impact of SHP2 inhibition on murine host immune cells and the tumor immune microenvironment in vivo using RMC-4550, a novel small-molecule allosteric inhibitor of SHP2. Oral daily administration of RMC-4550 significantly inhibited tumor growth in three syngeneic tumor models sensitive to checkpoint blockade. The inhibitory activity was comparable, and in some cases superior, to checkpoint inhibition. RMC-4550 did not inhibit growth in any of these cancer cell lines in vitro, suggesting that activity was not due to a tumor cell intrinsic antiproliferative effect. Rather, antitumor activity in vivo reflected modulation of murine host immune cell function. First, RMC-4550 did not inhibit tumor growth in immunocompromised Rag-2-deficient mice. Second, efficacy was significantly attenuated when CD8+T-cells were depleted in immunocompetent mice, suggesting that CD8+T-cells were important for tumor growth inhibition. Third, Shp2 inhibition had additive activity in combination with anti-CTLA4 or anti-PD-L1 treatment, resulting in complete tumor regression in some mice. Rechallenge studies also demonstrated the presence of immunologic memory induced by combination therapy. The additive activity with checkpoint blockade suggests an additional mechanism of action beyond inhibition of the checkpoint signal. Fourth, analysis of the immune landscape in the tumor microenvironment indeed revealed modulation of both adaptive and innate immune mechanisms. Similar to checkpoint blockade, RMC-4550 increased the frequency of CD8+T-cell infiltrates in tumors with a corresponding decrease in their PD-1 expression. In addition, Shp2 inhibition significantly shifted polarized macrophage populations by markedly increasing M1 and decreasing M2, effects not seen with anti-CTLA4 or anti-PD-L1. Collectively, these results suggest that SHP2 inhibition is not identical
由PTPN11编码的蛋白酪氨酸磷酸酶SHP2是一种已知的癌症亚群的致癌驱动因子,也是RTK-RAS-MAPK通路的中心信号节点。遗传和药理学证据支持SHP2在驱动依赖于一系列激活的rtk、某些RAS和BRAF突变以及NF1功能丧失突变的癌细胞增殖中的作用。相比之下,SHP2在抗肿瘤免疫中的作用尚未得到很好的证实。SHP2结合免疫细胞中调控受体上磷酸化的ITIM和ITAM结构域,多个报告证实了SHP2/PD-1的物理相互作用。最近有人提出SHP2通过CD28的直接去磷酸化来转导PD-1抑制检查点信号。在这项研究中,我们发现一种由PD-1 ITAM中两个酪氨酸磷酸化的9-mers序列组成的肽(通过PEG8连接物连接)可以激活纯化的SHP2酶。我们还证明,与检查点抑制剂一样,SHP2的变构抑制可以激活报告基因PD-1/PD-L1中nfat介导的基因表达。基于这些发现,我们利用新型SHP2小分子变构抑制剂rmmc -4550,评估了SHP2抑制对小鼠宿主免疫细胞和体内肿瘤免疫微环境的影响。每日口服rmmc -4550可显著抑制三种对检查点阻断敏感的同基因肿瘤模型的肿瘤生长。抑制活性与检查点抑制相当,在某些情况下优于检查点抑制。rmmc -4550在体外没有抑制任何这些癌细胞系的生长,表明活性不是由于肿瘤细胞固有的抗增殖作用。相反,体内抗肿瘤活性反映了小鼠宿主免疫细胞功能的调节。首先,rmmc -4550不能抑制免疫功能低下的rag -2缺陷小鼠的肿瘤生长。其次,当CD8+ t细胞在免疫功能正常的小鼠中被耗尽时,其疗效显著减弱,这表明CD8+ t细胞对肿瘤生长抑制很重要。第三,Shp2抑制与抗ctla4或抗pd - l1治疗联合具有加性活性,导致部分小鼠肿瘤完全消退。再挑战研究也证明了联合治疗引起的免疫记忆的存在。检查点阻断的附加活性表明,除了抑制检查点信号外,还有另一种作用机制。第四,对肿瘤微环境中的免疫景观的分析确实揭示了适应性和先天免疫机制的调节。与检查点阻断类似,RMC-4550增加了肿瘤中CD8+ t细胞浸润的频率,相应降低了PD-1的表达。此外,Shp2抑制通过显著增加M1和降低M2来显著改变极化巨噬细胞群,这是抗ctla4或抗pd - l1所没有的效果。总的来说,这些结果表明SHP2抑制与检查点阻断不同,代表了一种新的研究策略,可以同时利用两种抗肿瘤机制:直接抑制癌细胞生长和调节肿瘤微环境。含有对SHP2敏感的致癌驱动突变和对检查点抑制剂具有临床敏感性的肿瘤可能特别感兴趣。引文格式:Elsa Quintana, Kasia Mordec, Robert J. Nichols, David Wildes, Chris J. Schulze, Darienne R. Myers, Mallika Singh, Elena Koltun, Adrian Gill, Stephen Kelsey, Mark A Goldsmith, Jan A.M.史密斯。SHP2的变构抑制通过调节先天和适应性机制诱导pd -1敏感肿瘤的抗肿瘤免疫[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 A103。
{"title":"Abstract A103: Allosteric inhibition of SHP2 induces antitumor immunity in PD-1-sensitive tumors through modulation of both innate and adaptive mechanisms","authors":"E. Quintana, Kasia Mordec, R. Nichols, D. Wildes, C. Schulze, D. Myers, Mallika Singh, E. Koltun, A. Gill, S. Kelsey, M. Goldsmith, Jan Smith","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A103","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A103","url":null,"abstract":"The protein-tyrosine phosphatase SHP2, encoded by PTPN11, is a known oncogenic driver in a subset of cancers and a central signaling node in the RTK-RAS-MAPK pathway. Genetic and pharmacologic evidence supports a role for SHP2 in driving the proliferation of cancer cells dependent upon a range of activated RTKs, certain RAS and BRAF mutations, and NF1 loss of function mutations. In contrast, a role for SHP2 in antitumor immunity is not well established. SHP2 binds to phosphorylated ITIM and ITAM domains on regulatory receptors in immune cells and multiple reports have demonstrated a SHP2/PD-1 physical interaction. Recently it has been proposed that SHP2 transduces the PD-1 inhibitory checkpoint signal by direct de-phosphorylation of CD28. In this study we show that a peptide comprising two tyrosine phosphorylated 9-mers sequences from the PD-1 ITAM (connected by a PEG8 linker) can activate purified SHP2 enzyme. We also demonstrate that, like checkpoint inhibitors, allosteric inhibition of SHP2 activates NFAT-mediated gene expression in a reporter gene PD-1/PD-L1 bioassay. Based on these findings, we evaluated the impact of SHP2 inhibition on murine host immune cells and the tumor immune microenvironment in vivo using RMC-4550, a novel small-molecule allosteric inhibitor of SHP2. Oral daily administration of RMC-4550 significantly inhibited tumor growth in three syngeneic tumor models sensitive to checkpoint blockade. The inhibitory activity was comparable, and in some cases superior, to checkpoint inhibition. RMC-4550 did not inhibit growth in any of these cancer cell lines in vitro, suggesting that activity was not due to a tumor cell intrinsic antiproliferative effect. Rather, antitumor activity in vivo reflected modulation of murine host immune cell function. First, RMC-4550 did not inhibit tumor growth in immunocompromised Rag-2-deficient mice. Second, efficacy was significantly attenuated when CD8+T-cells were depleted in immunocompetent mice, suggesting that CD8+T-cells were important for tumor growth inhibition. Third, Shp2 inhibition had additive activity in combination with anti-CTLA4 or anti-PD-L1 treatment, resulting in complete tumor regression in some mice. Rechallenge studies also demonstrated the presence of immunologic memory induced by combination therapy. The additive activity with checkpoint blockade suggests an additional mechanism of action beyond inhibition of the checkpoint signal. Fourth, analysis of the immune landscape in the tumor microenvironment indeed revealed modulation of both adaptive and innate immune mechanisms. Similar to checkpoint blockade, RMC-4550 increased the frequency of CD8+T-cell infiltrates in tumors with a corresponding decrease in their PD-1 expression. In addition, Shp2 inhibition significantly shifted polarized macrophage populations by markedly increasing M1 and decreasing M2, effects not seen with anti-CTLA4 or anti-PD-L1. Collectively, these results suggest that SHP2 inhibition is not identical","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":"77546912","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}
引用次数: 0
Abstract A096: The potential role of fibroblast activation protein as a natural killer cell immune checkpoint 摘要:成纤维细胞活化蛋白作为自然杀伤细胞免疫检查点的潜在作用
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A096
Allison O'Connell, Shangzi Wang, L. Weiner
Fibroblast activation protein (FAP) is a type II transmembrane serine protease that functions as both a dipeptidyl peptidase and an endopeptidase. FAP is minimally expressed in normal pancreas but overexpressed in 90% of pancreatic ductal adenocarcinoma (PDAC) specimens. A meta-analysis of PDAC studies demonstrated elevated tumor FAP expression is associated with worse clinical outcomes. While immunotherapy offers remarkable results for certain cancer types, it has been largely ineffective in PDAC. This lack of efficacy may be attributed to the dense stromal fibrosis, comprised largely of pancreatic stellate cells (PSCs), that is characteristic of PDAC lesions. Here we demonstrate that human NK cell line (NK92) is activated by and kill PSCs. Upon direct contact with PSCs, NK92 cells upregulate FAP. FAP expression by NK92 cells is associated with an inactivation phenotype. Talabostat, a non-specific inhibitor of FAP, enhances NK92 killing of PSCs in vitro and enhances PDAC tumor clearance in vivo. This suggests that FAP may be a novel NK cell immune checkpoint that can be pharmacologically modulated to enhance NK cell antitumor activity. Citation Format: Allison O9Connell, Shangzi Wang, Louis M. Weiner. The potential role of fibroblast activation protein as a natural killer cell immune checkpoint [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 A096.
成纤维细胞活化蛋白(FAP)是一种II型跨膜丝氨酸蛋白酶,具有二肽基肽酶和内肽酶的功能。FAP在正常胰腺中表达极低,但在90%的胰腺导管腺癌(PDAC)标本中过表达。PDAC研究的荟萃分析表明,肿瘤FAP表达升高与较差的临床结果相关。虽然免疫疗法对某些类型的癌症提供了显著的效果,但它在PDAC中基本上无效。这种疗效的缺乏可能归因于密集的间质纤维化,主要由胰腺星状细胞(PSCs)组成,这是PDAC病变的特征。在这里,我们证明了人类NK细胞系(NK92)被PSCs激活并杀死。直接接触PSCs后,NK92细胞上调FAP。NK92细胞的FAP表达与失活表型相关。Talabostat是一种非特异性FAP抑制剂,在体外增强NK92对PSCs的杀伤作用,在体内增强PDAC的肿瘤清除率。这表明FAP可能是一种新的NK细胞免疫检查点,可以通过药理调节来增强NK细胞的抗肿瘤活性。引用格式:Allison O9Connell, Wang Shangzi, Louis M. Weiner。成纤维细胞活化蛋白作为自然杀伤细胞免疫检查点的潜在作用[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 A096。
{"title":"Abstract A096: The potential role of fibroblast activation protein as a natural killer cell immune checkpoint","authors":"Allison O'Connell, Shangzi Wang, L. Weiner","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A096","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A096","url":null,"abstract":"Fibroblast activation protein (FAP) is a type II transmembrane serine protease that functions as both a dipeptidyl peptidase and an endopeptidase. FAP is minimally expressed in normal pancreas but overexpressed in 90% of pancreatic ductal adenocarcinoma (PDAC) specimens. A meta-analysis of PDAC studies demonstrated elevated tumor FAP expression is associated with worse clinical outcomes. While immunotherapy offers remarkable results for certain cancer types, it has been largely ineffective in PDAC. This lack of efficacy may be attributed to the dense stromal fibrosis, comprised largely of pancreatic stellate cells (PSCs), that is characteristic of PDAC lesions. Here we demonstrate that human NK cell line (NK92) is activated by and kill PSCs. Upon direct contact with PSCs, NK92 cells upregulate FAP. FAP expression by NK92 cells is associated with an inactivation phenotype. Talabostat, a non-specific inhibitor of FAP, enhances NK92 killing of PSCs in vitro and enhances PDAC tumor clearance in vivo. This suggests that FAP may be a novel NK cell immune checkpoint that can be pharmacologically modulated to enhance NK cell antitumor activity. Citation Format: Allison O9Connell, Shangzi Wang, Louis M. Weiner. The potential role of fibroblast activation protein as a natural killer cell immune checkpoint [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 A096.","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":"74654476","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}
引用次数: 1
Abstract A098: SLC12A2 as a novel “brake” on immunogenic apoptotic cell clearance 摘要:SLC12A2作为免疫原性凋亡细胞清除的新“制动器”
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A098
Justin S. A. Perry, Sho Morioka, C. B. Medina, Michael H. Raymond, K. Ravichandran
Phagocytes face a key challenge during efferocytosis – they must engulf an apoptotic cell, often similar in size, while maintaining their own cell integrity, shape, and volume. Yet little is known about phagocyte volume homeostasis during apoptotic cell clearance. To this end, we performed an unbiased transcriptomics screen designed to reveal alterations of phagocyte gene expression after engulfment of apoptotic Jurkat lymphoma cells and identified several transcriptional programs distinctly modified in engulfing phagocytes. We identified a program involving cell volume regulation and potassium/chloride flux, the latter of which is thought to underlie immunogenic responses. Disruption of one member of this cell volume program, SLC12A2, resulted in “overeating” of apoptotic JurkaT-cells by individual phagocytes and a significant increase in cell size. Importantly, we demonstrate that SLC12A2-deficient phagocytes actively engulfing apoptotic JurkaT-cells led to a downregulation of the hallmark antiinflammatory efferocytosis program and robust upregulation of a proinflammatory program. In particular, we observed robust upregulation of immunogenic programs, including a type I interferon signature, in SLC12A2-deficient engulfing phagocytes that were not observed in phagocytes over-expressing the phosphatidylserine receptor TIM4, Mechanistically, we found that the chloride-sensing kinase WNK1 and downstream kinases OSR1 and SPAK function to regulate chloride flux into engulfing phagocytes via SLC12A2, and that this pathway regulates the apoptotic JurkaT-cell “overeating” observed in SLC12A2-deficient phagocytes. Collectively, these data elucidate a novel mechanism by which phagocytes protect themselves from potentially dangerous apoptotic cell-derived inflammatory ligands and suggest that SLC12A2 serves to actively suppress immunogenic responses by phagocytes and limits the rate of corpse uptake during efferocytosis. Citation Format: Justin Shaun Arnold Perry, Sho Morioka, Christopher Medina, Michael Raymond, Kodi Ravichandran. SLC12A2 as a novel “brake” on immunogenic apoptotic cell clearance [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 A098.
吞噬细胞在efferocytosis过程中面临着一个关键的挑战——它们必须吞噬凋亡细胞,通常在大小上相似,同时保持自身细胞的完整性、形状和体积。然而,在凋亡细胞清除过程中,对吞噬细胞体积稳态的了解甚少。为此,我们进行了一项无偏倚的转录组学筛选,旨在揭示吞噬细胞凋亡后吞噬细胞基因表达的变化,并确定了几个在吞噬细胞中明显修饰的转录程序。我们确定了一个涉及细胞体积调节和钾/氯化物通量的程序,后者被认为是免疫原性反应的基础。细胞体积程序的一个成员SLC12A2被破坏,导致凋亡的jurkat细胞被单个吞噬细胞“暴饮暴食”,细胞大小显著增加。重要的是,我们证明了slc12a2缺陷的吞噬细胞主动吞噬凋亡的jurkat细胞导致抗炎efferocytosis程序的下调和促炎程序的强烈上调。特别是,我们在SLC12A2缺陷吞噬细胞中观察到包括I型干扰素信号在内的免疫原性程序的强烈上调,而在过表达磷脂酰丝氨酸受体TIM4的吞噬细胞中没有观察到。从机制上讲,我们发现氯感激酶WNK1和下游激酶OSR1和SPAK通过SLC12A2调节氯通量进入吞噬细胞。该通路调节slc12a2缺陷吞噬细胞中jurkat细胞的“暴饮暴食”凋亡。总的来说,这些数据阐明了吞噬细胞保护自己免受潜在危险的凋亡细胞衍生炎症配体的新机制,并表明SLC12A2可积极抑制吞噬细胞的免疫原性反应,并限制efferocytosis期间尸体摄取的速率。引文格式:Justin Shaun Arnold Perry, Sho Morioka, Christopher Medina, Michael Raymond, Kodi Ravichandran。SLC12A2作为免疫原性凋亡细胞清除的新“制动器”[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 A098。
{"title":"Abstract A098: SLC12A2 as a novel “brake” on immunogenic apoptotic cell clearance","authors":"Justin S. A. Perry, Sho Morioka, C. B. Medina, Michael H. Raymond, K. Ravichandran","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A098","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A098","url":null,"abstract":"Phagocytes face a key challenge during efferocytosis – they must engulf an apoptotic cell, often similar in size, while maintaining their own cell integrity, shape, and volume. Yet little is known about phagocyte volume homeostasis during apoptotic cell clearance. To this end, we performed an unbiased transcriptomics screen designed to reveal alterations of phagocyte gene expression after engulfment of apoptotic Jurkat lymphoma cells and identified several transcriptional programs distinctly modified in engulfing phagocytes. We identified a program involving cell volume regulation and potassium/chloride flux, the latter of which is thought to underlie immunogenic responses. Disruption of one member of this cell volume program, SLC12A2, resulted in “overeating” of apoptotic JurkaT-cells by individual phagocytes and a significant increase in cell size. Importantly, we demonstrate that SLC12A2-deficient phagocytes actively engulfing apoptotic JurkaT-cells led to a downregulation of the hallmark antiinflammatory efferocytosis program and robust upregulation of a proinflammatory program. In particular, we observed robust upregulation of immunogenic programs, including a type I interferon signature, in SLC12A2-deficient engulfing phagocytes that were not observed in phagocytes over-expressing the phosphatidylserine receptor TIM4, Mechanistically, we found that the chloride-sensing kinase WNK1 and downstream kinases OSR1 and SPAK function to regulate chloride flux into engulfing phagocytes via SLC12A2, and that this pathway regulates the apoptotic JurkaT-cell “overeating” observed in SLC12A2-deficient phagocytes. Collectively, these data elucidate a novel mechanism by which phagocytes protect themselves from potentially dangerous apoptotic cell-derived inflammatory ligands and suggest that SLC12A2 serves to actively suppress immunogenic responses by phagocytes and limits the rate of corpse uptake during efferocytosis. Citation Format: Justin Shaun Arnold Perry, Sho Morioka, Christopher Medina, Michael Raymond, Kodi Ravichandran. SLC12A2 as a novel “brake” on immunogenic apoptotic cell clearance [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 A098.","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":"79906281","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}
引用次数: 1
Abstract A075: A protective role for group 3 innate lymphoid cells in colitis-associated colorectal cancer 3组先天淋巴样细胞在结肠炎相关结直肠癌中的保护作用
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A075
J. Goc, N. Bessman, S. Sahota, F. Laure, G. Putzel, D. Withers, J. Arthur, Manish A. Shah, Gregory F. Sonnenberg
Chronic inflammation is recognized as a causative factor in the development of cancer and recent paradigms suggest that microbe-driven chronic inflammation is causally associated with the development and progression of cancer in the colon. Further, recent studies in mice have implicated innate lymphoid cells (ILC) as a key cell population that regulates intestinal inflammation. Group 3 innate lymphoid cells (ILC3) are essential regulators of immunity, inflammation and tissue homeostasis in the intestine, yet their role in cancer remains poorly defined. Here, we identify that ILC3 are associated with both human and mouse colon tumors. Tumor-associated ILC3 are selectively localized within lymphoid aggregates and exhibit a unique phenotypic profile as compared with nonmalignant tissue. Critically, mice with a selective deletion of ILC3-specific MHCII exhibit a striking increased susceptibility to intestinal tissue damage and develop highly invasive and flat colorectal tumors. These data demonstrate a protective role for antigen-presenting ILC3 in the context of cancer development and progression in the intestine, and suggest that further interrogation may lead to the development of novel immunotherapeutic strategies in colon cancer. Citation Format: Jeremy Goc, Nick Bessman, Sheena Sahota, Flamar Anne Laure, Gregory Putzel, David Withers, Janelle Arthur, Manish Shah, Gregory Sonnenberg. A protective role for group 3 innate lymphoid cells in colitis-associated colorectal cancer [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 A075.
慢性炎症被认为是癌症发展的一个致病因素,最近的研究范式表明,微生物驱动的慢性炎症与结肠癌的发生和进展有因果关系。此外,最近对小鼠的研究表明,先天淋巴样细胞(ILC)是调节肠道炎症的关键细胞群。第3组先天淋巴样细胞(ILC3)是肠道中免疫、炎症和组织稳态的重要调节因子,但它们在癌症中的作用仍不明确。在这里,我们发现ILC3与人类和小鼠结肠肿瘤有关。与非恶性组织相比,肿瘤相关的ILC3选择性地定位于淋巴细胞聚集体中,并表现出独特的表型特征。关键的是,选择性缺失ilc3特异性MHCII的小鼠对肠道组织损伤的易感性显著增加,并发展为高度侵袭性和扁平的结直肠肿瘤。这些数据证明了抗原呈递ILC3在肠道癌症发生和进展中的保护作用,并表明进一步的研究可能导致结肠癌新的免疫治疗策略的发展。引文格式:Jeremy Goc, Nick Bessman, Sheena Sahota, Flamar Anne Laure, Gregory Putzel, David Withers, Janelle Arthur, Manish Shah, Gregory Sonnenberg。先天淋巴样细胞3组在结肠炎相关结直肠癌中的保护作用[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 A075。
{"title":"Abstract A075: A protective role for group 3 innate lymphoid cells in colitis-associated colorectal cancer","authors":"J. Goc, N. Bessman, S. Sahota, F. Laure, G. Putzel, D. Withers, J. Arthur, Manish A. Shah, Gregory F. Sonnenberg","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A075","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A075","url":null,"abstract":"Chronic inflammation is recognized as a causative factor in the development of cancer and recent paradigms suggest that microbe-driven chronic inflammation is causally associated with the development and progression of cancer in the colon. Further, recent studies in mice have implicated innate lymphoid cells (ILC) as a key cell population that regulates intestinal inflammation. Group 3 innate lymphoid cells (ILC3) are essential regulators of immunity, inflammation and tissue homeostasis in the intestine, yet their role in cancer remains poorly defined. Here, we identify that ILC3 are associated with both human and mouse colon tumors. Tumor-associated ILC3 are selectively localized within lymphoid aggregates and exhibit a unique phenotypic profile as compared with nonmalignant tissue. Critically, mice with a selective deletion of ILC3-specific MHCII exhibit a striking increased susceptibility to intestinal tissue damage and develop highly invasive and flat colorectal tumors. These data demonstrate a protective role for antigen-presenting ILC3 in the context of cancer development and progression in the intestine, and suggest that further interrogation may lead to the development of novel immunotherapeutic strategies in colon cancer. Citation Format: Jeremy Goc, Nick Bessman, Sheena Sahota, Flamar Anne Laure, Gregory Putzel, David Withers, Janelle Arthur, Manish Shah, Gregory Sonnenberg. A protective role for group 3 innate lymphoid cells in colitis-associated colorectal cancer [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 A075.","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":"89630847","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}
引用次数: 0
Abstract A106: Higher numbers of cancer stem cells in the peripheral blood of children with B-ALL after chemotherapy A106:化疗后B-ALL患儿外周血中肿瘤干细胞数量增加
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A106
M. Salem, M. Attia, S. Abdou, Abdel-Aziz A Zidan, Mona F Zidan
Background: Acute lymphocytic leukemia (ALL) is biologically and clinically considered as a heterogeneous neoplasm of lymphoid progenitor cells in the bone marrow (BM). 15- 20 % of children with ALL who achieve an initial remission, will show relapse. One potential mechanism behind this relapse could be the emergence of cancer stem cells (CSCs), which are considered the driving force of tumorigenesis due to their ability of self-renewal as well as the emergence of immune regulatory cells including myeloid-derived suppressor cells (MDSCs) and regulatory T-cells (Treg). Aim: The main aim of this study was to analyze the numbers of CSCs and correlate these numbers with the numbers of blasT-cells, MDSCs and Treg cells in children with B-ALL before and after induction of chemotherapy. Materials and Methods: CSCs were defined as CD45dimCD19+CD10+CD34+CD38-, MDSCs were defined as Lin-HLA-DR-CD33+CD11b+ and Treg cells were defined as CD4+CD25+CD127-. The frequencies of these cells were analyzed in the peripheral blood of B-ALL patients before (n= 10) and after (n= 10) induction of chemotherapy using flow cytometry. Results: Significant increases in the numbers of CSCs were shown in B-ALL patients after induction of chemotherapy as compared to newly diagnosed patients (7.6± 8.3 vs. 2.7± 2.4, P Citation Format: Mohamed Labib Salem, Mohamed Attia, Said Abdou, Abdel-Aziz A. Zidan, Mona F. Zidan. Higher numbers of cancer stem cells in the peripheral blood of children with B-ALL after chemotherapy [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 A106.
背景:急性淋巴细胞白血病(Acute lymphocytic leukemia, ALL)在生物学和临床上被认为是一种骨髓淋巴样祖细胞(BM)的异质性肿瘤。15- 20%的ALL患儿在最初得到缓解后会复发。这种复发背后的一个潜在机制可能是癌症干细胞(CSCs)的出现,由于其自我更新的能力以及包括髓源性抑制细胞(MDSCs)和调节性t细胞(Treg)在内的免疫调节细胞的出现,CSCs被认为是肿瘤发生的驱动力。目的:本研究的主要目的是分析B-ALL患儿在诱导化疗前后CSCs的数量及其与blasT-cells、MDSCs和Treg细胞数量的关系。材料与方法:CSCs定义为CD45dimCD19+CD10+CD34+CD38-, MDSCs定义为Lin-HLA-DR-CD33+CD11b+, Treg细胞定义为CD4+CD25+CD127-。用流式细胞术分析B-ALL患者化疗前(n= 10)和诱导化疗后(n= 10)外周血中这些细胞的频率。结果:与新诊断的患者相比,B-ALL患者诱导化疗后的CSCs数量显著增加(7.6±8.3比2.7±2.4),P引用格式:Mohamed Labib Salem, Mohamed Attia, Said Abdou, Abdel-Aziz A. Zidan, Mona F. Zidan。化疗后B-ALL患儿外周血中肿瘤干细胞数量增加[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 A106。
{"title":"Abstract A106: Higher numbers of cancer stem cells in the peripheral blood of children with B-ALL after chemotherapy","authors":"M. Salem, M. Attia, S. Abdou, Abdel-Aziz A Zidan, Mona F Zidan","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A106","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A106","url":null,"abstract":"Background: Acute lymphocytic leukemia (ALL) is biologically and clinically considered as a heterogeneous neoplasm of lymphoid progenitor cells in the bone marrow (BM). 15- 20 % of children with ALL who achieve an initial remission, will show relapse. One potential mechanism behind this relapse could be the emergence of cancer stem cells (CSCs), which are considered the driving force of tumorigenesis due to their ability of self-renewal as well as the emergence of immune regulatory cells including myeloid-derived suppressor cells (MDSCs) and regulatory T-cells (Treg). Aim: The main aim of this study was to analyze the numbers of CSCs and correlate these numbers with the numbers of blasT-cells, MDSCs and Treg cells in children with B-ALL before and after induction of chemotherapy. Materials and Methods: CSCs were defined as CD45dimCD19+CD10+CD34+CD38-, MDSCs were defined as Lin-HLA-DR-CD33+CD11b+ and Treg cells were defined as CD4+CD25+CD127-. The frequencies of these cells were analyzed in the peripheral blood of B-ALL patients before (n= 10) and after (n= 10) induction of chemotherapy using flow cytometry. Results: Significant increases in the numbers of CSCs were shown in B-ALL patients after induction of chemotherapy as compared to newly diagnosed patients (7.6± 8.3 vs. 2.7± 2.4, P Citation Format: Mohamed Labib Salem, Mohamed Attia, Said Abdou, Abdel-Aziz A. Zidan, Mona F. Zidan. Higher numbers of cancer stem cells in the peripheral blood of children with B-ALL after chemotherapy [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 A106.","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":"87968135","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}
引用次数: 0
Abstract A121: Single-cell transcriptomic analyses reveal heterogeneity of vascular endothelial cells in cancer models 摘要:单细胞转录组学分析揭示了肿瘤模型中血管内皮细胞的异质性
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A121
Yu Zhu, Nicole Salazar, Kevin F. Brulois, E. Butcher
The immune system has demonstrated promising potential as a therapeutic target in the treatment of certain cancer types, yet in many tumors the efficacy of immunotherapies is still extremely poor. One major obstacle is the limited access of immune cells to the tumor tissue. The tumor vasculature plays critical roles in regulating the recruitment of anti-tumor immune cells and the delivery of immunotherapies to the tumor sites. These immune processes require functional specialization of endothelial cells (ECs). However, there is a major gap in our understanding of the diversity, molecular specialization, and developmental relationships of tumor-associated EC subsets. To understand the heterogeneity of tumor-associated endothelium, we performed single-cell RNA sequencing (scRNAseq) analyses on ECs isolated from mouse cancer models. scRNAseq identified several subsets of endothelium in the tumor tissue, including ECs that compose the artery, capillary, and post-capillary venules. Interestingly, within the capillary endothelial compartment, we identified an EC subset that resembles tip cells in sprouting angiogenesis and demonstrates molecular signature of stem or progenitor cells. These putative progenitor cells are dramatically expanded in tumors compared to normal tissues, and comprise both quiescent and proliferative populations. The quiescent ECs express high levels of genes coding for antiapoptotic proteins, extracellular matrix molecules, immune checkpoint molecules, monocyte/macrophage recruiting cytokines, and stem cell signaling components, such as Notch4. On the other hand, the proliferative tip-like ECs express genes coding for cyclins and cyclin-dependent kinases, DNA damage repair molecules, necroptosis machinery, and epigenetic regulators. Taken together, these data define the heterogeneity of tumor-associated endothelial cells, and reveal a putative progenitor population that gives rise to the tumor vasculature and could be targeted to improve the efficacy of immunotherapies. Citation Format: Yu Zhu, Nicole Salazar, Kevin Brulois, Eugene Butcher. Single-cell transcriptomic analyses reveal heterogeneity of vascular endothelial cells in cancer models [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 A121.
免疫系统作为治疗某些类型癌症的靶点已经显示出巨大的潜力,然而在许多肿瘤中,免疫疗法的疗效仍然非常差。一个主要的障碍是免疫细胞进入肿瘤组织的途径有限。肿瘤脉管系统在调节抗肿瘤免疫细胞的募集和向肿瘤部位输送免疫疗法方面起着至关重要的作用。这些免疫过程需要内皮细胞(ECs)的功能特化。然而,我们对肿瘤相关EC亚群的多样性、分子专门化和发育关系的理解存在重大差距。为了了解肿瘤相关内皮的异质性,我们对从小鼠癌症模型中分离的内皮细胞进行了单细胞RNA测序(scRNAseq)分析。scRNAseq在肿瘤组织中发现了几个内皮亚群,包括构成动脉、毛细血管和毛细血管后小静脉的内皮细胞。有趣的是,在毛细血管内皮腔室中,我们发现了一个EC亚群,它类似于发芽血管生成中的尖端细胞,并显示出干细胞或祖细胞的分子特征。与正常组织相比,这些假定的祖细胞在肿瘤中急剧扩增,并包括静止和增殖的群体。静止的ECs表达高水平的基因编码抗凋亡蛋白、细胞外基质分子、免疫检查点分子、单核/巨噬细胞募集细胞因子和干细胞信号传导成分,如Notch4。另一方面,增殖尖端样ECs表达细胞周期蛋白和细胞周期蛋白依赖性激酶、DNA损伤修复分子、坏死坏死机制和表观遗传调节因子的编码基因。综上所述,这些数据定义了肿瘤相关内皮细胞的异质性,并揭示了一个推定的祖细胞群,该祖细胞群可以产生肿瘤血管,并可以靶向提高免疫治疗的疗效。引文格式:Yu Zhu, Nicole Salazar, Kevin Brulois, Eugene Butcher。单细胞转录组学分析揭示了肿瘤模型中血管内皮细胞的异质性[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 A121。
{"title":"Abstract A121: Single-cell transcriptomic analyses reveal heterogeneity of vascular endothelial cells in cancer models","authors":"Yu Zhu, Nicole Salazar, Kevin F. Brulois, E. Butcher","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A121","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A121","url":null,"abstract":"The immune system has demonstrated promising potential as a therapeutic target in the treatment of certain cancer types, yet in many tumors the efficacy of immunotherapies is still extremely poor. One major obstacle is the limited access of immune cells to the tumor tissue. The tumor vasculature plays critical roles in regulating the recruitment of anti-tumor immune cells and the delivery of immunotherapies to the tumor sites. These immune processes require functional specialization of endothelial cells (ECs). However, there is a major gap in our understanding of the diversity, molecular specialization, and developmental relationships of tumor-associated EC subsets. To understand the heterogeneity of tumor-associated endothelium, we performed single-cell RNA sequencing (scRNAseq) analyses on ECs isolated from mouse cancer models. scRNAseq identified several subsets of endothelium in the tumor tissue, including ECs that compose the artery, capillary, and post-capillary venules. Interestingly, within the capillary endothelial compartment, we identified an EC subset that resembles tip cells in sprouting angiogenesis and demonstrates molecular signature of stem or progenitor cells. These putative progenitor cells are dramatically expanded in tumors compared to normal tissues, and comprise both quiescent and proliferative populations. The quiescent ECs express high levels of genes coding for antiapoptotic proteins, extracellular matrix molecules, immune checkpoint molecules, monocyte/macrophage recruiting cytokines, and stem cell signaling components, such as Notch4. On the other hand, the proliferative tip-like ECs express genes coding for cyclins and cyclin-dependent kinases, DNA damage repair molecules, necroptosis machinery, and epigenetic regulators. Taken together, these data define the heterogeneity of tumor-associated endothelial cells, and reveal a putative progenitor population that gives rise to the tumor vasculature and could be targeted to improve the efficacy of immunotherapies. Citation Format: Yu Zhu, Nicole Salazar, Kevin Brulois, Eugene Butcher. Single-cell transcriptomic analyses reveal heterogeneity of vascular endothelial cells in cancer models [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 A121.","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":"86922529","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}
引用次数: 0
Abstract A078: Dissecting the myeloid lineage in human gliomas 摘要:人胶质瘤髓系的解剖
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A078
Claudia Z. Han, S. Duttke, Zhengyu Ouyang, S. Preissl, J. Schlachetzki, Alexander Nott, Conor Fitzpatrick, Carolyn O’Connor, N. Coufal, Mihir Gupta, D. Gonda, M. Levy, Ben-Haim Sharona, Barba David, J. Ciacci, A. Khalessi, Clark C. Chen, Bing Ren, C. Glass
The immune cell composition of the tumor microenvironment can be a decisive factor for tumor pathogenesis. Gliomas are tumors that develop from the glial cells of the brain and spinal cord and make up to 30% of all brain tumors. In gliomas, microglia and infiltrating macrophages can comprise up to 30 to 50% of total tumor-associated cells. Increased CD68 staining, a marker of microglia/macrophages, in adult gliomas is positively associated with histologic tumor grade. Despite the accumulated evidence substantiating a critical role for microglia and infiltrating macrophages in gliomagenesis, little is known is about the molecular mechanisms driving microglial contribution to tumor growth and whether microglia/macrophages are therapeutic targets in both low- and high-grade gliomas. Despite microglia sharing common properties with other tissue-resident macrophages, they express hundreds of genes at higher levels compared to other tissue-resident macrophages, many of which are influenced by the brain micro-environment. Additionally, engraftment of bone-marrow derived cells into the central nervous system fails to produce microglia identical to yolk sac-derived microglia at the transcriptional level. Hence, in any inflammatory context, including cancer, an interesting question arises: how does each population contribute to the pathogenesis and/or resolution of inflammation? To elucidate the role(s) of microglia/macrophages in gliomas, we isolated the myeloid fraction from primary pediatric and adult low-grade and high-grade gliomas using flow cytometry. By integrating bulk and single-cell transcriptome analysis, we find significant inter- and intratumoral heterogeneity within the myeloid population. Additionally, we find evidence for tumor environment-dependent gene change. In combination with ongoing comparative analysis of the corresponding epigenetic landscapes of the myeloid populations, we seek to decipher how the tumor microenvironment reprograms the transcription factor network in microglia/macrophages to generate tumor-promoting cells. Citation Format: Claudia Z. Han, Sascha H. Duttke, Zhengyu Ouyang, Sebastian Preissl, Johannes C.M. Schlachetzki, Alexander Nott, Conor Fitzpatrick, Carolyn O9Connor, Nicole G. Coufal, Mihir Gupta, David D. Gonda, Michael L. Levy, Ben-Haim Sharona, Barba David, Joseph D. Ciacci, Alexander A. Khalessi, Clark C. Chen, Bing Ren, Christopher K. Glass. Dissecting the myeloid lineage in human gliomas [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 A078.
肿瘤微环境的免疫细胞组成可能是肿瘤发病的决定性因素。神经胶质瘤是由大脑和脊髓的神经胶质细胞发展而来的肿瘤,占所有脑肿瘤的30%。在胶质瘤中,小胶质细胞和浸润性巨噬细胞可占肿瘤相关细胞总数的30%至50%。成人胶质瘤中CD68染色(小胶质细胞/巨噬细胞的标志物)的增加与组织学肿瘤分级呈正相关。尽管越来越多的证据表明小胶质细胞和浸润性巨噬细胞在胶质瘤形成中起着关键作用,但关于驱动小胶质细胞促进肿瘤生长的分子机制以及小胶质细胞/巨噬细胞是否是低级别和高级别胶质瘤的治疗靶点,我们知之甚少。尽管小胶质细胞与其他组织驻留巨噬细胞具有共同的特性,但与其他组织驻留巨噬细胞相比,它们表达的数百个基因水平更高,其中许多基因受脑微环境的影响。此外,骨髓源性细胞移植到中枢神经系统后,在转录水平上不能产生与卵黄囊源性小胶质细胞相同的小胶质细胞。因此,在任何炎症背景下,包括癌症,一个有趣的问题出现了:每个人群如何促进炎症的发病和/或解决?为了阐明小胶质细胞/巨噬细胞在胶质瘤中的作用,我们使用流式细胞术分离了原发性小儿和成人低级别和高级别胶质瘤的髓系部分。通过整合整体和单细胞转录组分析,我们发现髓系人群中存在显著的肿瘤间和肿瘤内异质性。此外,我们还发现了肿瘤环境相关基因变化的证据。结合正在进行的髓系群体相应表观遗传景观的比较分析,我们试图破译肿瘤微环境如何重编程小胶质细胞/巨噬细胞中的转录因子网络以产生促肿瘤细胞。引文格式:Claudia Z. Han, Sascha H. Duttke,欧阳正宇,Sebastian Preissl, Johannes C.M. Schlachetzki, Alexander Nott, Conor Fitzpatrick, Carolyn O9Connor, Nicole G. Coufal, Mihir Gupta, David D. Gonda, Michael L. Levy, Ben-Haim Sharona, Barba David, Joseph D. Ciacci, Alexander A. Khalessi, Clark C. Chen, Bing Ren, Christopher K. Glass。人胶质瘤的髓系谱系解剖[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 A078。
{"title":"Abstract A078: Dissecting the myeloid lineage in human gliomas","authors":"Claudia Z. Han, S. Duttke, Zhengyu Ouyang, S. Preissl, J. Schlachetzki, Alexander Nott, Conor Fitzpatrick, Carolyn O’Connor, N. Coufal, Mihir Gupta, D. Gonda, M. Levy, Ben-Haim Sharona, Barba David, J. Ciacci, A. Khalessi, Clark C. Chen, Bing Ren, C. Glass","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A078","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A078","url":null,"abstract":"The immune cell composition of the tumor microenvironment can be a decisive factor for tumor pathogenesis. Gliomas are tumors that develop from the glial cells of the brain and spinal cord and make up to 30% of all brain tumors. In gliomas, microglia and infiltrating macrophages can comprise up to 30 to 50% of total tumor-associated cells. Increased CD68 staining, a marker of microglia/macrophages, in adult gliomas is positively associated with histologic tumor grade. Despite the accumulated evidence substantiating a critical role for microglia and infiltrating macrophages in gliomagenesis, little is known is about the molecular mechanisms driving microglial contribution to tumor growth and whether microglia/macrophages are therapeutic targets in both low- and high-grade gliomas. Despite microglia sharing common properties with other tissue-resident macrophages, they express hundreds of genes at higher levels compared to other tissue-resident macrophages, many of which are influenced by the brain micro-environment. Additionally, engraftment of bone-marrow derived cells into the central nervous system fails to produce microglia identical to yolk sac-derived microglia at the transcriptional level. Hence, in any inflammatory context, including cancer, an interesting question arises: how does each population contribute to the pathogenesis and/or resolution of inflammation? To elucidate the role(s) of microglia/macrophages in gliomas, we isolated the myeloid fraction from primary pediatric and adult low-grade and high-grade gliomas using flow cytometry. By integrating bulk and single-cell transcriptome analysis, we find significant inter- and intratumoral heterogeneity within the myeloid population. Additionally, we find evidence for tumor environment-dependent gene change. In combination with ongoing comparative analysis of the corresponding epigenetic landscapes of the myeloid populations, we seek to decipher how the tumor microenvironment reprograms the transcription factor network in microglia/macrophages to generate tumor-promoting cells. Citation Format: Claudia Z. Han, Sascha H. Duttke, Zhengyu Ouyang, Sebastian Preissl, Johannes C.M. Schlachetzki, Alexander Nott, Conor Fitzpatrick, Carolyn O9Connor, Nicole G. Coufal, Mihir Gupta, David D. Gonda, Michael L. Levy, Ben-Haim Sharona, Barba David, Joseph D. Ciacci, Alexander A. Khalessi, Clark C. Chen, Bing Ren, Christopher K. Glass. Dissecting the myeloid lineage in human gliomas [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 A078.","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":"86401626","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}
引用次数: 0
期刊
Tackling the Tumor Microenvironment: Beyond T-cells
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