首页 > 最新文献

Checkpoints and Immunomodulation最新文献

英文 中文
Abstract A21: Activation of 4-1BB on liver myeloid cells triggers hepatitis via an interleukin-27 dependent pathway 摘要:4-1BB在肝髓细胞上的激活通过白细胞介素-27依赖途径触发肝炎
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A21
A. Jaiswal, T. Bartkowiak, C. Ager, Renee L Chin, Chao-Hsien Chen, Pratha Budhani, Midan Ai, M. Reilley, Manu Sebastian, D. Hong, M. Curran
Purpose: Agonist antibodies targeting the T cell co-stimulatory receptor 4-1BB (CD137) are among the most effective immunotherapeutic agents across multiple preclinical models of cancer. In the clinic, however, development of these agents has been stymied by dose-limiting liver toxicity. Lack of knowledge of the mechanisms underlying this toxicity has limited the potential to separate 4-1BB agonist driven anti-tumor immunity from hepatotoxicity. Experimental Design: The capacity of 4-1BB agonist antibodies to induce liver toxicity was investigated in immune competent mice, with or without co-administration of checkpoint blockade, via measurement of serum transaminase levels, through imaging of liver immune infiltrates, and via qualitative and quantitative assessment of liver myeloid and T cells via flow cytometry. Knockout mice were used to clarify the contribution of specific cell subsets, cytokines and chemokines. Results: We find that activation of 4-1BB on liver myeloid cells is essential to initiate hepatitis. Once activated, these cells produce interleukin-27, which is required for liver toxicity. CD8 T cells infiltrate the liver in response to this myeloid activation and mediate tissue damage triggering transaminase elevation. FoxP3+ regulatory T cells limit liver damage and their removal dramatically exacerbates 4-1BB agonist hepatitis. Co-administration of CTLA-4 blockade ameliorates transaminase elevation, whereas PD-1 blockade exacerbates it. Loss of the chemokine receptor CCR2 blocks 4-1BB agonist hepatitis without diminishing tumor-specific immunity against B16 melanoma. Conclusions: 4-1BB agonist antibodies trigger hepatitis via activation of myeloid cells to produce Interleukin-27. Co-administration of CTLA-4 and/or CCR2 blockade may minimize hepatitis but yields equal or greater antitumor immunity. Citation Format: Ashvin R. Jaiswal, Todd Bartkowiak, Casey R. Ager, Renee Chin, Chao Hsien Chen, Pratha Budhani, Midan Ai, Matthew J. Reilley, Manu M. Sebastian, David Hong, Michael A. Curran. Activation of 4-1BB on liver myeloid cells triggers hepatitis via an interleukin-27 dependent pathway [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A21.
目的:靶向T细胞共刺激受体4-1BB (CD137)的激动剂抗体是多种癌症临床前模型中最有效的免疫治疗药物之一。然而,在临床上,这些药物的开发一直受到剂量限制性肝毒性的阻碍。缺乏对这种毒性机制的了解限制了将4-1BB激动剂驱动的抗肿瘤免疫从肝毒性中分离出来的可能性。实验设计:通过测量血清转氨酶水平、肝脏免疫浸润成像以及流式细胞术对肝髓细胞和T细胞进行定性和定量评估,在免疫正常的小鼠中研究了4-1BB激动剂抗体诱导肝毒性的能力,无论是否联合使用检查点阻断。用敲除小鼠来阐明特定细胞亚群、细胞因子和趋化因子的作用。结果:我们发现4-1BB对肝髓细胞的激活是引发肝炎的必要条件。一旦被激活,这些细胞就会产生白细胞介素-27,这是肝毒性所必需的。CD8 T细胞浸润肝脏,响应骨髓活化,介导组织损伤,引发转氨酶升高。FoxP3+调节性T细胞限制肝损伤,其去除可显著加重4-1BB激动性肝炎。CTLA-4阻断可改善转氨酶升高,而PD-1阻断可加重转氨酶升高。趋化因子受体CCR2的缺失可阻断4-1BB激动剂肝炎,但不降低针对B16黑色素瘤的肿瘤特异性免疫。结论:4-1BB激动剂抗体通过激活髓细胞产生白细胞介素-27触发肝炎。同时使用CTLA-4和/或CCR2阻断剂可以减少肝炎,但产生相同或更大的抗肿瘤免疫。引文格式:Ashvin R. Jaiswal, Todd Bartkowiak, Casey R. Ager, Renee Chin, Chao Hsien Chen, Pratha Budhani, Midan Ai, Matthew J. Reilley, Manu M. Sebastian, David Hong, Michael A. Curran肝髓细胞上4-1BB的激活通过白细胞介素-27依赖途径触发肝炎[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫学杂志,2018;6(9增刊):摘要nr A21。
{"title":"Abstract A21: Activation of 4-1BB on liver myeloid cells triggers hepatitis via an interleukin-27 dependent pathway","authors":"A. Jaiswal, T. Bartkowiak, C. Ager, Renee L Chin, Chao-Hsien Chen, Pratha Budhani, Midan Ai, M. Reilley, Manu Sebastian, D. Hong, M. Curran","doi":"10.1158/2326-6074.TUMIMM17-A21","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A21","url":null,"abstract":"Purpose: Agonist antibodies targeting the T cell co-stimulatory receptor 4-1BB (CD137) are among the most effective immunotherapeutic agents across multiple preclinical models of cancer. In the clinic, however, development of these agents has been stymied by dose-limiting liver toxicity. Lack of knowledge of the mechanisms underlying this toxicity has limited the potential to separate 4-1BB agonist driven anti-tumor immunity from hepatotoxicity. Experimental Design: The capacity of 4-1BB agonist antibodies to induce liver toxicity was investigated in immune competent mice, with or without co-administration of checkpoint blockade, via measurement of serum transaminase levels, through imaging of liver immune infiltrates, and via qualitative and quantitative assessment of liver myeloid and T cells via flow cytometry. Knockout mice were used to clarify the contribution of specific cell subsets, cytokines and chemokines. Results: We find that activation of 4-1BB on liver myeloid cells is essential to initiate hepatitis. Once activated, these cells produce interleukin-27, which is required for liver toxicity. CD8 T cells infiltrate the liver in response to this myeloid activation and mediate tissue damage triggering transaminase elevation. FoxP3+ regulatory T cells limit liver damage and their removal dramatically exacerbates 4-1BB agonist hepatitis. Co-administration of CTLA-4 blockade ameliorates transaminase elevation, whereas PD-1 blockade exacerbates it. Loss of the chemokine receptor CCR2 blocks 4-1BB agonist hepatitis without diminishing tumor-specific immunity against B16 melanoma. Conclusions: 4-1BB agonist antibodies trigger hepatitis via activation of myeloid cells to produce Interleukin-27. Co-administration of CTLA-4 and/or CCR2 blockade may minimize hepatitis but yields equal or greater antitumor immunity. Citation Format: Ashvin R. Jaiswal, Todd Bartkowiak, Casey R. Ager, Renee Chin, Chao Hsien Chen, Pratha Budhani, Midan Ai, Matthew J. Reilley, Manu M. Sebastian, David Hong, Michael A. Curran. Activation of 4-1BB on liver myeloid cells triggers hepatitis via an interleukin-27 dependent pathway [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A21.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73902274","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 A29: Multimeric anti-human OX40 induces robust immune responses A29:多聚体抗人OX40诱导强大的免疫应答
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A29
Shravan Madireddi, Yanli Yang, Sherry H Yeh, Chen Gu, Patricia L. Sanchez, Randall J. Brezski, H. Chiu, H. Erickson, R. Cubas, G. Lazar, J. Kim
Agonistic antibodies to co-stimulatory members of the TNF Receptor Superfamily (TNFRSF) have demonstrated impressive antitumor immune responses in murine tumor models and are currently under investigation in the clinic. In order to induce efficient co-stimulatory signaling, the majority of TNFRSF members require supermolecular cluster formation on the immune cell surface. Recent studies have revealed that agonistic antibodies achieve this in part by Fcγ receptor (FcγR) mediated cross-linking, enabling antibodies to cluster the TNFRSF member expressed on the opposing immune cell. Agonistic antibodies to OX40 (CD134; TNFRSF4), which primarily target T cells are currently in clinical trials for cancer immunotherapy. However, as OX40 needs clustering for efficient signaling, these antibodies can only function in microenvironments where FcγR bearing cells are abundant. To overcome this limitation, we generated multimeric anti-human-OX40 antibodies, which cluster OX40 independent of FcγR mediated cross-linking. In vitro, we found that multimeric anti-OX40 induces robust signaling independent of FcγR. Compared to the bivalent antibodies, multimeric anti-OX40 generated superior immune responses in vaccination and tumor settings, in vivo. Together, multimeric antibody platforms have important implications for effective targeting of OX40 and possibly other TNFRSF members. Citation Format: Shravan Madireddi, Yanli Yang, Sherry Yeh, Chen Gu, Patricia L. Sanchez, Randall Brezski, Henry Chiu, Hans Erickson, Rafael A. Cubas, Gregory Lazar, Jeong Kim. Multimeric anti-human OX40 induces robust immune responses [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A29.
针对TNF受体超家族(TNFRSF)共刺激成员的激动抗体在小鼠肿瘤模型中显示出令人印象深刻的抗肿瘤免疫反应,目前正在临床研究中。为了诱导有效的共刺激信号,大多数TNFRSF成员需要在免疫细胞表面形成超分子簇。最近的研究表明,激动抗体部分通过Fcγ受体(Fcγ r)介导的交联实现这一目标,使抗体能够聚集在对立免疫细胞上表达的TNFRSF成员。OX40 (CD134)激动抗体;TNFRSF4),主要靶向T细胞,目前正处于癌症免疫治疗的临床试验中。然而,由于OX40需要聚集才能有效地传递信号,这些抗体只能在fc - γ - r承载细胞丰富的微环境中发挥作用。为了克服这一限制,我们产生了多聚体抗人OX40抗体,该抗体不依赖于FcγR介导的交联。在体外,我们发现多聚体抗ox40诱导了不依赖于FcγR的强大信号。与二价抗体相比,多聚体抗ox40在疫苗接种和肿瘤环境中产生了更好的体内免疫应答。总之,多聚抗体平台对于有效靶向OX40和其他TNFRSF成员具有重要意义。引文格式:Shravan Madireddi, Yanli Yang, Sherry Yeh, Chen Gu, Patricia L. Sanchez, Randall Brezski, Henry Chiu, Hans Erickson, Rafael A. Cubas, Gregory Lazar, Jeong Kim。多聚体抗人OX40诱导强大的免疫应答[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫,2018;6(9增刊):摘要nr A29。
{"title":"Abstract A29: Multimeric anti-human OX40 induces robust immune responses","authors":"Shravan Madireddi, Yanli Yang, Sherry H Yeh, Chen Gu, Patricia L. Sanchez, Randall J. Brezski, H. Chiu, H. Erickson, R. Cubas, G. Lazar, J. Kim","doi":"10.1158/2326-6074.TUMIMM17-A29","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A29","url":null,"abstract":"Agonistic antibodies to co-stimulatory members of the TNF Receptor Superfamily (TNFRSF) have demonstrated impressive antitumor immune responses in murine tumor models and are currently under investigation in the clinic. In order to induce efficient co-stimulatory signaling, the majority of TNFRSF members require supermolecular cluster formation on the immune cell surface. Recent studies have revealed that agonistic antibodies achieve this in part by Fcγ receptor (FcγR) mediated cross-linking, enabling antibodies to cluster the TNFRSF member expressed on the opposing immune cell. Agonistic antibodies to OX40 (CD134; TNFRSF4), which primarily target T cells are currently in clinical trials for cancer immunotherapy. However, as OX40 needs clustering for efficient signaling, these antibodies can only function in microenvironments where FcγR bearing cells are abundant. To overcome this limitation, we generated multimeric anti-human-OX40 antibodies, which cluster OX40 independent of FcγR mediated cross-linking. In vitro, we found that multimeric anti-OX40 induces robust signaling independent of FcγR. Compared to the bivalent antibodies, multimeric anti-OX40 generated superior immune responses in vaccination and tumor settings, in vivo. Together, multimeric antibody platforms have important implications for effective targeting of OX40 and possibly other TNFRSF members. Citation Format: Shravan Madireddi, Yanli Yang, Sherry Yeh, Chen Gu, Patricia L. Sanchez, Randall Brezski, Henry Chiu, Hans Erickson, Rafael A. Cubas, Gregory Lazar, Jeong Kim. Multimeric anti-human OX40 induces robust immune responses [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A29.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"75313068","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 A28: Tumor cell intrinsic BPTF inhibits NK cell activity and the abundance of natural cytotoxicity receptor co-ligands 摘要:肿瘤细胞内禀BPTF抑制NK细胞活性和天然细胞毒性受体共配体的丰度
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A28
Kimberly Mayes, Zeinab Elsayed, Aiman S Alhazmi, M. Waters, Suehyb G. Alkhatib, Mark Roberts, Carolyn Song, Kristen Peterson, Vivian Chan, Nikhil Ailaney, Pumoli Malapati, T. Blevins, Berislav Lisnić, C. Dumur, Joseph W. Landry
Using syngeneic BALB/c mouse breast cancer models, we show that the chromatin remodeling subunit bromodomain PHD finger transcription factor (BPTF) suppresses natural killer (NK) cell antitumor activity in the tumor microenvironment (TME). In culture, BPTF suppresses direct natural cytotoxicity receptor (NCR) mediated NK cell cytolytic activity to mouse and human cancer cell lines, demonstrating conserved functions. Blocking mouse NCR1 in vivo rescues BPTF KD tumor weights, demonstrating its importance for the control of tumor growth. We discovered that BPTF occupies heparanase (Hpse) regulatory elements, activating its expression. Increased heparanase activity results in reduced cell surface abundance of the NCR co-ligands: heparan sulfate proteoglycans (HSPGs). Using gain and loss of function approaches we show that elevated heparanase levels suppress NK cell cytolytic activity to tumor cells in culture. These results suggest that BPTF activates heparanase expression, which in turn reduces cell surface HSPGs and NCR co-ligands, inhibiting NK cell activity. Furthermore, gene expression data from human breast cancer tumors shows that elevated BPTF expression correlates with reduced antitumor immune cell signatures, supporting conserved roles for BPTF in suppressing antitumor immunity. Conditional BPTF depletion in established mouse breast tumors enhances antitumor immunity, suggesting that inhibiting BPTF could provide a novel immunotherapy. Citation Format: Kimberly Mayes, Zeinab Elsayed, Aiman Alhazmi, Michael Waters, Suehyb Alkhatib, Mark Roberts, Carolyn Song, Kristen Peterson, Vivian Chan, Nikhil Ailaney, Pumoli Malapati, Tana Blevins, Berislav Lisnic, Catherine Dumur, Joseph Landry. Tumor cell intrinsic BPTF inhibits NK cell activity and the abundance of natural cytotoxicity receptor co-ligands [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A28.
利用同基因BALB/c小鼠乳腺癌模型,我们发现染色质重塑亚基bromodomain PHD手指转录因子(BPTF)在肿瘤微环境(TME)中抑制自然杀伤细胞(NK)抗肿瘤活性。在培养中,BPTF抑制直接天然细胞毒性受体(NCR)介导的NK细胞对小鼠和人类癌细胞的细胞溶解活性,显示出保守的功能。在体内阻断小鼠NCR1可挽救BPTF KD肿瘤重量,证明其对控制肿瘤生长的重要性。我们发现BPTF占据肝素酶(Hpse)调控元件,激活其表达。肝素酶活性的增加导致NCR共配体:硫酸肝素蛋白聚糖(HSPGs)的细胞表面丰度降低。使用增益和功能损失的方法,我们表明升高的肝素酶水平抑制NK细胞对肿瘤细胞的细胞溶解活性。这些结果表明,BPTF激活肝素酶表达,从而减少细胞表面HSPGs和NCR共配体,抑制NK细胞活性。此外,来自人类乳腺癌肿瘤的基因表达数据显示,BPTF表达升高与抗肿瘤免疫细胞特征降低相关,支持BPTF在抑制抗肿瘤免疫中的保守作用。在已建立的小鼠乳腺肿瘤中,条件性BPTF耗竭可增强抗肿瘤免疫,表明抑制BPTF可能提供一种新的免疫治疗方法。引文格式:Kimberly Mayes, Zeinab Elsayed, Aiman Alhazmi, Michael Waters, Suehyb Alkhatib, Mark Roberts, Carolyn Song, Kristen Peterson, Vivian Chan, Nikhil Ailaney, Pumoli Malapati, Tana Blevins, Berislav Lisnic, Catherine Dumur, Joseph Landry。肿瘤细胞内禀BPTF抑制NK细胞活性和天然细胞毒性受体共配体的丰度[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫,2018;6(9增刊):摘要nr A28。
{"title":"Abstract A28: Tumor cell intrinsic BPTF inhibits NK cell activity and the abundance of natural cytotoxicity receptor co-ligands","authors":"Kimberly Mayes, Zeinab Elsayed, Aiman S Alhazmi, M. Waters, Suehyb G. Alkhatib, Mark Roberts, Carolyn Song, Kristen Peterson, Vivian Chan, Nikhil Ailaney, Pumoli Malapati, T. Blevins, Berislav Lisnić, C. Dumur, Joseph W. Landry","doi":"10.1158/2326-6074.TUMIMM17-A28","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A28","url":null,"abstract":"Using syngeneic BALB/c mouse breast cancer models, we show that the chromatin remodeling subunit bromodomain PHD finger transcription factor (BPTF) suppresses natural killer (NK) cell antitumor activity in the tumor microenvironment (TME). In culture, BPTF suppresses direct natural cytotoxicity receptor (NCR) mediated NK cell cytolytic activity to mouse and human cancer cell lines, demonstrating conserved functions. Blocking mouse NCR1 in vivo rescues BPTF KD tumor weights, demonstrating its importance for the control of tumor growth. We discovered that BPTF occupies heparanase (Hpse) regulatory elements, activating its expression. Increased heparanase activity results in reduced cell surface abundance of the NCR co-ligands: heparan sulfate proteoglycans (HSPGs). Using gain and loss of function approaches we show that elevated heparanase levels suppress NK cell cytolytic activity to tumor cells in culture. These results suggest that BPTF activates heparanase expression, which in turn reduces cell surface HSPGs and NCR co-ligands, inhibiting NK cell activity. Furthermore, gene expression data from human breast cancer tumors shows that elevated BPTF expression correlates with reduced antitumor immune cell signatures, supporting conserved roles for BPTF in suppressing antitumor immunity. Conditional BPTF depletion in established mouse breast tumors enhances antitumor immunity, suggesting that inhibiting BPTF could provide a novel immunotherapy. Citation Format: Kimberly Mayes, Zeinab Elsayed, Aiman Alhazmi, Michael Waters, Suehyb Alkhatib, Mark Roberts, Carolyn Song, Kristen Peterson, Vivian Chan, Nikhil Ailaney, Pumoli Malapati, Tana Blevins, Berislav Lisnic, Catherine Dumur, Joseph Landry. Tumor cell intrinsic BPTF inhibits NK cell activity and the abundance of natural cytotoxicity receptor co-ligands [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A28.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"77354976","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 IA32: Improving checkpoint blockade for lymphoma with Flt3L-primed in situ vaccination IA32: flt3l引物原位疫苗改善淋巴瘤检查点阻断
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-IA32
J. Brody
Checkpoint blockade therapy of cancer has had tremendous impact; still, only a subset of patients experience remissions. One hypothesis is that resistant tumors lack sufficient somatic mutational burden and thus, targetable tumor-associated antigens (TAA). To the contrary, we described that Hodgkin’s lymphoma, despite a high anti-PD1 response rate (~65-87%), has significantly fewer mutations (Reichel et al., Blood 2015) than highly mutated tumors, such as lung cancer (anti-PD1 response rate ~20%). Recent, seminal work confirms that the presence of dendritic cells (DC) capable of cross-presenting TAA correlates with tumor immunogenicity (Spranger et al., Proc Natl Acad Sci USA 2016). Our hypothesis is that checkpoint blockade is limited by the tolerogenic microenvironment, specifically, suboptimal cross-presentation of TAA by suitably activated dendritic cells (DC). If so, then checkpoint blockade will be potentiated by optimal recruitment, loading, and activation of cross-presenting DC at the tumor site. We and others have recently found that tumors—including lymphomas—evade immune clearance by active exclusion of DC. Despite progress in mechanistic understanding of this immune evasion, we still lack means to modulate the phenomenon and facilitate T-cell entry and destruction of tumors. We developed an early-phase trial (funded by Damon Runyon CRF) testing a unique, rationally designed in situ vaccine (ISV) comprising 1) Flt3L to recruit DC, 2) radiotherapy (XRT) to load Flt3L-mobilized DC with TAA, and 3) Toll-like receptor agonist (TLRa) to activate TAA-loaded DC for cross-presentation. Strikingly, we observed partial and complete systemic tumor regressions, including distant and untreated tumors, improving months after therapy, and even elimination of malignant B cells with sparing of healthy B cells, suggesting a systemic antitumor immune response. The trial is based on our preclinical dat,a which recapitulate the clinical findings and also show that the ISV cure rate (~40%) increases markedly by combination with PD1 blockade (80-90%). These data prompted a new trial combining ISV with PD1 blockade opening in 2018. Though PD1/PDL1 blockade is potentiated by in situ vaccination, it is possible that there may be other, even more effective checkpoints for combination. We have developed two separate, complementary screening approaches using a novel EGFP-specific, murine CD8 T cell to enrich or probe for novel tumor-expressed (“PD-L1-like”) checkpoint molecules and have identified candidates for validation. Ultimately, the screening can be performed in vivo in the context of various immunotherapies, including in situ vaccination, stem cell transplantation, or PD-1 blockade. Citation Format: Joshua D. Brody. Improving checkpoint blockade for lymphoma with Flt3L-primed in situ vaccination [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol
检查点阻断疗法对癌症产生了巨大的影响;尽管如此,只有一小部分患者的病情得到缓解。一种假设是,耐药肿瘤缺乏足够的体细胞突变负担,因此缺乏可靶向的肿瘤相关抗原(TAA)。相反,我们描述了霍奇金淋巴瘤,尽管抗pd1反应率高(~65-87%),但突变明显少于高突变的肿瘤,如肺癌(抗pd1反应率~20%)(Reichel等,Blood 2015)。最近,开创性的工作证实,能够交叉呈递TAA的树突状细胞(DC)的存在与肿瘤免疫原性相关(Spranger等人,Proc Natl Acad Sci USA 2016)。我们的假设是检查点阻断受到耐受性微环境的限制,特别是适当激活的树突状细胞(DC)的TAA交叉呈现不理想。如果是这样,那么检查点阻断将通过肿瘤部位交叉呈递DC的最佳招募、装载和激活而增强。我们和其他人最近发现,肿瘤(包括淋巴瘤)通过主动排除DC来逃避免疫清除。尽管对这种免疫逃避的机制理解有所进展,但我们仍然缺乏调节这种现象和促进t细胞进入和破坏肿瘤的手段。我们开展了一项早期试验(由Damon Runyon CRF资助),测试了一种独特的、合理设计的原位疫苗(ISV),该疫苗包括:1)Flt3L招募DC, 2)放疗(XRT)将Flt3L动员的DC装载TAA,以及3)toll样受体激动剂(TLRa)激活装载TAA的DC进行交叉呈递。引人注目的是,我们观察到部分和完全的全身肿瘤消退,包括远处和未经治疗的肿瘤,治疗后几个月改善,甚至消除了恶性B细胞,保留了健康B细胞,这表明全身抗肿瘤免疫反应。该试验基于我们的临床前数据,这些数据概括了临床结果,并表明与PD1阻断剂(80-90%)联合使用ISV治愈率(~40%)显着提高。这些数据促使ISV与PD1阻断剂的新试验于2018年开始。虽然原位疫苗接种增强了PD1/PDL1阻断,但可能存在其他更有效的联合检查点。我们已经开发了两种独立的、互补的筛选方法,使用一种新的egfp特异性小鼠CD8 T细胞来富集或探测新的肿瘤表达(“pd - l1样”)检查点分子,并确定了候选分子进行验证。最终,筛选可以在各种免疫疗法的背景下在体内进行,包括原位疫苗接种,干细胞移植或PD-1阻断。引用格式:Joshua D. Brody。flt3l引物原位疫苗改善淋巴瘤检查点阻断[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫学杂志,2018;6(9增刊):摘要nr - IA32。
{"title":"Abstract IA32: Improving checkpoint blockade for lymphoma with Flt3L-primed in situ vaccination","authors":"J. Brody","doi":"10.1158/2326-6074.TUMIMM17-IA32","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-IA32","url":null,"abstract":"Checkpoint blockade therapy of cancer has had tremendous impact; still, only a subset of patients experience remissions. One hypothesis is that resistant tumors lack sufficient somatic mutational burden and thus, targetable tumor-associated antigens (TAA). To the contrary, we described that Hodgkin’s lymphoma, despite a high anti-PD1 response rate (~65-87%), has significantly fewer mutations (Reichel et al., Blood 2015) than highly mutated tumors, such as lung cancer (anti-PD1 response rate ~20%). Recent, seminal work confirms that the presence of dendritic cells (DC) capable of cross-presenting TAA correlates with tumor immunogenicity (Spranger et al., Proc Natl Acad Sci USA 2016). Our hypothesis is that checkpoint blockade is limited by the tolerogenic microenvironment, specifically, suboptimal cross-presentation of TAA by suitably activated dendritic cells (DC). If so, then checkpoint blockade will be potentiated by optimal recruitment, loading, and activation of cross-presenting DC at the tumor site. We and others have recently found that tumors—including lymphomas—evade immune clearance by active exclusion of DC. Despite progress in mechanistic understanding of this immune evasion, we still lack means to modulate the phenomenon and facilitate T-cell entry and destruction of tumors. We developed an early-phase trial (funded by Damon Runyon CRF) testing a unique, rationally designed in situ vaccine (ISV) comprising 1) Flt3L to recruit DC, 2) radiotherapy (XRT) to load Flt3L-mobilized DC with TAA, and 3) Toll-like receptor agonist (TLRa) to activate TAA-loaded DC for cross-presentation. Strikingly, we observed partial and complete systemic tumor regressions, including distant and untreated tumors, improving months after therapy, and even elimination of malignant B cells with sparing of healthy B cells, suggesting a systemic antitumor immune response. The trial is based on our preclinical dat,a which recapitulate the clinical findings and also show that the ISV cure rate (~40%) increases markedly by combination with PD1 blockade (80-90%). These data prompted a new trial combining ISV with PD1 blockade opening in 2018. Though PD1/PDL1 blockade is potentiated by in situ vaccination, it is possible that there may be other, even more effective checkpoints for combination. We have developed two separate, complementary screening approaches using a novel EGFP-specific, murine CD8 T cell to enrich or probe for novel tumor-expressed (“PD-L1-like”) checkpoint molecules and have identified candidates for validation. Ultimately, the screening can be performed in vivo in the context of various immunotherapies, including in situ vaccination, stem cell transplantation, or PD-1 blockade. Citation Format: Joshua D. Brody. Improving checkpoint blockade for lymphoma with Flt3L-primed in situ vaccination [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"80230240","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 A19: PD-1 modulation promotes antitumor immunity by improving metabolic fitness of both PD-1+ and PD-1- CD8+ T cells in the tumor 摘要:PD-1调节通过改善肿瘤中PD-1+和PD-1- CD8+ T细胞的代谢适合度来促进抗肿瘤免疫
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A19
Kristen E. Pauken, Vikram R. Juneja, P. Sage, M. LaFleur, J. Kuchroo, A. Ringel, N. Ron-Harel, Seth Maleri, G. Freeman, Nicolas Chevrier, M. Haigis, A. Sharpe
Although PD-1 pathway inhibitors are revolutionizing cancer treatment, the mechanisms by which PD-1 regulates anti-tumor immunity are not fully understood. Following subcutaneous transplantation of MC38 adenocarcinoma tumor cells into mice, we show that complete loss of PD-1 selectively on CD8+ T cells improved metabolic activity and functions in the tumor microenvironment (TME). Since clinically PD-1 inhibitors likely act on T cells post-priming, we next deleted PD-1 after initial priming and restricted deletion to roughly 50% of cells. Loss of PD-1 led to T cell-intrinsic boosts in metabolism and CD8+ T cells that lost PD-1 after priming preferentially formed anti-tumor memory cells, suggesting PD-1 antagonizes memory formation. Unexpectedly, there was also a bystander effect that improved functions of PD-1 expressing CD8+ T cells in the TME. These data suggest that complete loss of PD-1 is not necessary for optimal tumor immunity, and that enhancing the functions of a subset of CD8+ T cells can promote an antitumor microenvironment and immunologic memory. Citation Format: Kristen E. Pauken, Vikram R. Juneja, Peter T. Sage, Martin W. LaFleur, Juhi R. Kuchroo, Alison Ringel, Noga Ron-Harel, Seth P. Maleri, Gordon J. Freeman, Nicolas Chevrier, Marcia C. Haigis, Arlene H. Sharpe. PD-1 modulation promotes antitumor immunity by improving metabolic fitness of both PD-1+ and PD-1- CD8+ T cells in the tumor [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A19.
尽管PD-1途径抑制剂正在彻底改变癌症治疗,但PD-1调节抗肿瘤免疫的机制尚不完全清楚。将MC38腺癌肿瘤细胞皮下移植到小鼠体内后,我们发现CD8+ T细胞上PD-1的选择性完全丧失改善了肿瘤微环境(TME)中的代谢活性和功能。由于临床上PD-1抑制剂可能在T细胞启动后起作用,因此我们在初始启动后删除PD-1,并将删除限制在大约50%的细胞中。PD-1缺失导致T细胞内在代谢增强,启动后缺失PD-1的CD8+ T细胞优先形成抗肿瘤记忆细胞,提示PD-1拮抗记忆形成。出乎意料的是,还存在一种旁观者效应,可以改善TME中表达CD8+ T细胞的PD-1功能。这些数据表明,PD-1的完全丧失并不是最佳肿瘤免疫的必要条件,增强CD8+ T细胞亚群的功能可以促进抗肿瘤微环境和免疫记忆。引文格式:Kristen E. Pauken, Vikram R. Juneja, Peter T. Sage, Martin W. LaFleur, Juhi R. Kuchroo, Alison Ringel, Noga Ron-Harel, Seth P. Maleri, Gordon J. Freeman, Nicolas Chevrier, Marcia C. Haigis, Arlene H. SharpePD-1调节通过改善肿瘤中PD-1+和PD-1- CD8+ T细胞的代谢适合度来促进抗肿瘤免疫[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫学杂志,2018;6(9增刊):摘要nr - A19。
{"title":"Abstract A19: PD-1 modulation promotes antitumor immunity by improving metabolic fitness of both PD-1+ and PD-1- CD8+ T cells in the tumor","authors":"Kristen E. Pauken, Vikram R. Juneja, P. Sage, M. LaFleur, J. Kuchroo, A. Ringel, N. Ron-Harel, Seth Maleri, G. Freeman, Nicolas Chevrier, M. Haigis, A. Sharpe","doi":"10.1158/2326-6074.TUMIMM17-A19","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A19","url":null,"abstract":"Although PD-1 pathway inhibitors are revolutionizing cancer treatment, the mechanisms by which PD-1 regulates anti-tumor immunity are not fully understood. Following subcutaneous transplantation of MC38 adenocarcinoma tumor cells into mice, we show that complete loss of PD-1 selectively on CD8+ T cells improved metabolic activity and functions in the tumor microenvironment (TME). Since clinically PD-1 inhibitors likely act on T cells post-priming, we next deleted PD-1 after initial priming and restricted deletion to roughly 50% of cells. Loss of PD-1 led to T cell-intrinsic boosts in metabolism and CD8+ T cells that lost PD-1 after priming preferentially formed anti-tumor memory cells, suggesting PD-1 antagonizes memory formation. Unexpectedly, there was also a bystander effect that improved functions of PD-1 expressing CD8+ T cells in the TME. These data suggest that complete loss of PD-1 is not necessary for optimal tumor immunity, and that enhancing the functions of a subset of CD8+ T cells can promote an antitumor microenvironment and immunologic memory. Citation Format: Kristen E. Pauken, Vikram R. Juneja, Peter T. Sage, Martin W. LaFleur, Juhi R. Kuchroo, Alison Ringel, Noga Ron-Harel, Seth P. Maleri, Gordon J. Freeman, Nicolas Chevrier, Marcia C. Haigis, Arlene H. Sharpe. PD-1 modulation promotes antitumor immunity by improving metabolic fitness of both PD-1+ and PD-1- CD8+ T cells in the tumor [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A19.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"89185950","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 A17: Bromodomain and extraterminal proteins regulate PD-L1/PD-1 signaling in breast cancer. 摘要:溴域和外膜蛋白在乳腺癌中调控PD-L1/PD-1信号传导。
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A17
G. Andrieu, G. Denis
Introduction: Breast tumors are massively infiltrated immune cells of both lymphoid and myeloid origin that exert an anti-tumor pressure to limit cancer progression. Tumor cells can escape immune clearance by expressing several molecules that constitute the immune checkpoints. Antibodies targeting the PD-L1/PD-1 pathway are being evaluated clinically for several cancers and show early success. However, the realization that not all patients respond well to immunotherapy suggests other modalities could be combined to improve efficacy. The bromodomain and extraterminal (BET) proteins BRD2, BRD3 and BRD4 are epigenetically acting co-regulators of transcription, and are critical for cancer proliferation and metastasis. Little is known about the molecular mechanisms that regulate PD-L1 and PD-1 expression. BRD4 is known to bind directly to the PD-L1 promoter; its targeting suppresses PD-L1 expression while increasing CD8+ T cell activity to limit progression in ovarian tumor models. Hypotheses: BET proteins regulate PD-L1 and PD-L2 expression in breast tumors and PD-1 in CD8+ T cells. BET proteins are effectors of proinflammatory cytokine signaling, regulating PD-L1/PD-1 signaling in the breast tumor microenvironment. Methods: We used different cellular models of breast cancer. We also collected human peripheral blood mononuclear cells matched with mammary adipose tissue from mammoplasty patients at Boston Medical Center to assess expression of immune checkpoint proteins in the breast microenvironment. Pan-BET protein inhibition was performed by JQ1 treatment. Alternatively, BET proteins were individually and specifically depleted by siRNA. PD-1 and PD-L1 expression were determined by qRT-PCR and flow cytometry. Cytokines and chemokines present in the blood or secreted by the mammary adipose tissue were profiled by multiplexed antibody capture assay. Results: BET proteins regulate PD-L1 expression by breast cancer cells. BRD2 and BRD4 expression correlate with PD-1 in activated circulating T cells and BET inhibition ablates activation-induced PD-1 expression. Co-culture of breast cancer cells with activated circulating T cells enhances expression of immune checkpoint proteins in both compartments. BET protein inhibition rescues these phenotypes. Conditioned media from mammary adipose tissue increase PD-1 in PBMCs. Peripheral and mammary adipose tissue inflammatory signatures associate with PD-1 expression in PBMCs, suggesting that local inflammation in the breast tumor microenvironment contributes to diminishing anti-tumor immune responses. Conclusion: BET proteins regulate the PD-L1/PD-1 pathway in breast cancer. Next-generation BET protein inhibitors may combine well with PD-1 or PD-L1-targeted immunotherapies in difficult-to-treat cancers. Personalized BET profiles could inform individual patient responses to BET proteins and immune checkpoint inhibitors. Therapeutic approaches that treat the microenvironment should be leveraged to maximize efficacy
导读:乳腺肿瘤是淋巴和髓源性免疫细胞大量浸润,发挥抗肿瘤压力来限制肿瘤进展。肿瘤细胞可以通过表达构成免疫检查点的几个分子来逃避免疫清除。针对PD-L1/PD-1通路的抗体正在临床评估几种癌症,并显示出早期的成功。然而,认识到并非所有患者对免疫治疗反应良好,表明可以结合其他方式来提高疗效。溴结构域和外端(BET)蛋白BRD2、BRD3和BRD4是表观遗传学上的转录共同调节因子,对癌症的增殖和转移至关重要。调控PD-L1和PD-1表达的分子机制尚不清楚。已知BRD4可直接结合PD-L1启动子;其靶向性抑制PD-L1表达,同时增加CD8+ T细胞活性,以限制卵巢肿瘤模型的进展。假设:BET蛋白调节乳腺肿瘤中PD-L1和PD-L2的表达以及CD8+ T细胞中PD-1的表达。BET蛋白是促炎细胞因子信号转导的效应器,调节乳腺肿瘤微环境中PD-L1/PD-1信号转导。方法:采用不同的乳腺癌细胞模型。我们还收集了波士顿医疗中心乳房成型术患者与乳腺脂肪组织匹配的人外周血单个核细胞,以评估乳房微环境中免疫检查点蛋白的表达。JQ1对Pan-BET蛋白进行抑制。或者,BET蛋白被siRNA单独和特异性地耗尽。采用qRT-PCR和流式细胞术检测PD-1和PD-L1的表达。用多重抗体捕获法分析了血液中或乳腺脂肪组织分泌的细胞因子和趋化因子。结果:BET蛋白调控PD-L1在乳腺癌细胞中的表达。在活化的循环T细胞中,BRD2和BRD4的表达与PD-1相关,BET抑制可抑制活化诱导的PD-1表达。乳腺癌细胞与活化的循环T细胞共培养可增强两个区室中免疫检查点蛋白的表达。BET蛋白抑制挽救了这些表型。来自乳腺脂肪组织的条件培养基增加PBMCs中的PD-1。外周和乳腺脂肪组织炎症特征与PBMCs中PD-1的表达相关,表明乳腺肿瘤微环境中的局部炎症有助于降低抗肿瘤免疫反应。结论:BET蛋白在乳腺癌中调控PD-L1/PD-1通路。下一代BET蛋白抑制剂可能与PD-1或pd - l1靶向免疫疗法在难以治疗的癌症中很好地结合。个性化BET档案可以告知个体患者对BET蛋白和免疫检查点抑制剂的反应。应该利用治疗微环境的治疗方法来最大化检查点抑制剂方法对特定癌症的疗效。引文格式:Guillaume Andrieu, Gerald V. Denis。溴结构域和外膜蛋白调控PD-L1/PD-1在乳腺癌中的信号传导。[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫学杂志,2018;6(9增刊):摘要nr A17。
{"title":"Abstract A17: Bromodomain and extraterminal proteins regulate PD-L1/PD-1 signaling in breast cancer.","authors":"G. Andrieu, G. Denis","doi":"10.1158/2326-6074.TUMIMM17-A17","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A17","url":null,"abstract":"Introduction: Breast tumors are massively infiltrated immune cells of both lymphoid and myeloid origin that exert an anti-tumor pressure to limit cancer progression. Tumor cells can escape immune clearance by expressing several molecules that constitute the immune checkpoints. Antibodies targeting the PD-L1/PD-1 pathway are being evaluated clinically for several cancers and show early success. However, the realization that not all patients respond well to immunotherapy suggests other modalities could be combined to improve efficacy. The bromodomain and extraterminal (BET) proteins BRD2, BRD3 and BRD4 are epigenetically acting co-regulators of transcription, and are critical for cancer proliferation and metastasis. Little is known about the molecular mechanisms that regulate PD-L1 and PD-1 expression. BRD4 is known to bind directly to the PD-L1 promoter; its targeting suppresses PD-L1 expression while increasing CD8+ T cell activity to limit progression in ovarian tumor models. Hypotheses: BET proteins regulate PD-L1 and PD-L2 expression in breast tumors and PD-1 in CD8+ T cells. BET proteins are effectors of proinflammatory cytokine signaling, regulating PD-L1/PD-1 signaling in the breast tumor microenvironment. Methods: We used different cellular models of breast cancer. We also collected human peripheral blood mononuclear cells matched with mammary adipose tissue from mammoplasty patients at Boston Medical Center to assess expression of immune checkpoint proteins in the breast microenvironment. Pan-BET protein inhibition was performed by JQ1 treatment. Alternatively, BET proteins were individually and specifically depleted by siRNA. PD-1 and PD-L1 expression were determined by qRT-PCR and flow cytometry. Cytokines and chemokines present in the blood or secreted by the mammary adipose tissue were profiled by multiplexed antibody capture assay. Results: BET proteins regulate PD-L1 expression by breast cancer cells. BRD2 and BRD4 expression correlate with PD-1 in activated circulating T cells and BET inhibition ablates activation-induced PD-1 expression. Co-culture of breast cancer cells with activated circulating T cells enhances expression of immune checkpoint proteins in both compartments. BET protein inhibition rescues these phenotypes. Conditioned media from mammary adipose tissue increase PD-1 in PBMCs. Peripheral and mammary adipose tissue inflammatory signatures associate with PD-1 expression in PBMCs, suggesting that local inflammation in the breast tumor microenvironment contributes to diminishing anti-tumor immune responses. Conclusion: BET proteins regulate the PD-L1/PD-1 pathway in breast cancer. Next-generation BET protein inhibitors may combine well with PD-1 or PD-L1-targeted immunotherapies in difficult-to-treat cancers. Personalized BET profiles could inform individual patient responses to BET proteins and immune checkpoint inhibitors. Therapeutic approaches that treat the microenvironment should be leveraged to maximize efficacy","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"89422518","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 A30: Ron kinase inhibition to improve immunotherapy for breast cancer metastasis 摘要A30: Ron激酶抑制改善乳腺癌转移的免疫治疗
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A30
Shuping Lai, Atakan Ekiz, A. Welm
Metastasis is the cause of death for nearly all types of cancer, including breast cancer. An exciting new area of research in metastatic breast cancer centers on immune therapy. Although new immune checkpoint blockade therapies have provided benefit for a fraction of patients tested so far, the majority of patients still do not respond to these drugs. A better understanding of how the immune system can be harnessed against metastatic breast cancer is required in order to improve patient outcomes in this area. We previously discovered that macrophage Ron receptor tyrosine kinase promotes breast cancer metastasis by inhibiting CD8+ cytotoxic T lymphocyte activity. We hypothesized that dual blockade of Ron activity and existing immune checkpoint molecules would unleash a more effective CD8+ T cell response to control or eliminate metastatic breast cancer. Our strategy would simultaneously disable tumor-mediated immune evasion mechanisms on both the innate and adaptive immune systems. To test the potential of combination therapy, the Ron inhibitor BMS-777607 and/or the immune checkpoint blocking agents anti-PD1 or anti-CTLA4, were administered in our mouse mammary tumor model to examine the effects on breast tumor progression. To examine Ron-specific effects of BMS-777607, we also examined the effect of genetic deletion of host Ron signaling activity in combination with immunotherapy. The anti-tumor immune response was comprehensively examined by multi-color flow cytometry and immunohistochemical staining for tumor-infiltrating lymphocytes (TILs). Our data suggest that the combination of Ron inhibition with immune checkpoint blockade may be an effective therapy in breast cancer. Citation Format: Shu Chin Alicia Lai, Atakan Ekiz, Alana Welm. Ron kinase inhibition to improve immunotherapy for breast cancer metastasis [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A30.
转移几乎是所有癌症的死亡原因,包括乳腺癌。转移性乳腺癌的一个令人兴奋的新研究领域集中在免疫治疗上。尽管到目前为止,新的免疫检查点阻断疗法已经为一小部分患者提供了益处,但大多数患者仍然对这些药物没有反应。为了改善这一领域的患者预后,需要更好地了解如何利用免疫系统来对抗转移性乳腺癌。我们之前发现巨噬细胞Ron受体酪氨酸激酶通过抑制CD8+细胞毒性T淋巴细胞活性促进乳腺癌转移。我们假设Ron活性和现有免疫检查点分子的双重阻断将释放更有效的CD8+ T细胞反应来控制或消除转移性乳腺癌。我们的策略将同时在先天和适应性免疫系统中禁用肿瘤介导的免疫逃避机制。为了测试联合治疗的潜力,我们在小鼠乳腺肿瘤模型中给予Ron抑制剂BMS-777607和/或抗pd1或抗ctla4免疫检查点阻断剂,以检查对乳腺肿瘤进展的影响。为了研究BMS-777607的非特异性作用,我们还研究了基因缺失与免疫治疗联合对宿主Ron信号活性的影响。采用多色流式细胞术和肿瘤浸润淋巴细胞(TILs)免疫组化染色全面检测抗肿瘤免疫应答。我们的数据表明,Ron抑制与免疫检查点阻断联合可能是一种有效的乳腺癌治疗方法。引文格式:Shu Chin Alicia Lai, Atakan Ekiz, Alana Welm。Ron激酶抑制改善乳腺癌转移的免疫治疗[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫学杂志,2018;6(9增刊):摘要nr A30。
{"title":"Abstract A30: Ron kinase inhibition to improve immunotherapy for breast cancer metastasis","authors":"Shuping Lai, Atakan Ekiz, A. Welm","doi":"10.1158/2326-6074.TUMIMM17-A30","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A30","url":null,"abstract":"Metastasis is the cause of death for nearly all types of cancer, including breast cancer. An exciting new area of research in metastatic breast cancer centers on immune therapy. Although new immune checkpoint blockade therapies have provided benefit for a fraction of patients tested so far, the majority of patients still do not respond to these drugs. A better understanding of how the immune system can be harnessed against metastatic breast cancer is required in order to improve patient outcomes in this area. We previously discovered that macrophage Ron receptor tyrosine kinase promotes breast cancer metastasis by inhibiting CD8+ cytotoxic T lymphocyte activity. We hypothesized that dual blockade of Ron activity and existing immune checkpoint molecules would unleash a more effective CD8+ T cell response to control or eliminate metastatic breast cancer. Our strategy would simultaneously disable tumor-mediated immune evasion mechanisms on both the innate and adaptive immune systems. To test the potential of combination therapy, the Ron inhibitor BMS-777607 and/or the immune checkpoint blocking agents anti-PD1 or anti-CTLA4, were administered in our mouse mammary tumor model to examine the effects on breast tumor progression. To examine Ron-specific effects of BMS-777607, we also examined the effect of genetic deletion of host Ron signaling activity in combination with immunotherapy. The anti-tumor immune response was comprehensively examined by multi-color flow cytometry and immunohistochemical staining for tumor-infiltrating lymphocytes (TILs). Our data suggest that the combination of Ron inhibition with immune checkpoint blockade may be an effective therapy in breast cancer. Citation Format: Shu Chin Alicia Lai, Atakan Ekiz, Alana Welm. Ron kinase inhibition to improve immunotherapy for breast cancer metastasis [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A30.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"90106856","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 A16: Epstein-Barr virus encoded EBNA2 alters immune checkpoint PD-L1 expression by downregulating miR-34a in B cell lymphomas 摘要:eb病毒编码的EBNA2通过下调miR-34a改变B细胞淋巴瘤免疫检查点PD-L1的表达
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A16
Anastasiadou Eleni, Stroopinsky Dina, Stella Alimperti, Alan L Jiao, A. Pyzer, Claudia Cipitelli, M. Severa, Christopher S. Chen, S. Uccini, D. Avigan, A. Faggioni, P. Trivedi, F. Slack
Cancer cells subvert host immune surveillance by altering immune checkpoint (IC) proteins. Indeed, Epstein-Barr virus (EBV) positive Hodgkin’s lymphoma and gastric adenocarcinomas have higher Programmed Cell Death Ligand, PD-L1, expression. However, how EBV alters ICs in the context of its preferred host, the B lymphocyte and lymphomas, is unknown. Here, we used Burkitt lymphoma (BL), diffuse large B cell lymphomas (DLBCL) and their EBV infected or EBNA2 transfected derivatives to address this question. EBV latency III cells expressed high levels of PD-L1. In a DLBCL model, we found that EBNA2 but not LMP1 is sufficient to induce PD-L1. The upregulation of PD-L1 was confirmed in estrogen inducible EBNA2 carrying B lymphoma cells. Clinical samples from DLBCL patients showed that EBV infected, latency III cases expressed high levels of PD-L1. The PD-L1 targeting oncosuppressor miR-34a was downregulated in EBNA2 transfected cells. miR-34a reconstitution in EBNA2 expressing DLBCL reduced PD-L1 expression and increased their immunogenicity in 2D mixed lymphocyte reactions (MLR) and 3D microfluidic chip based MLRs. Given the importance of PD-L1 inhibition in immunotherapy and miR-34a dysregulation in cancers, our findings may have important implications for diagnosis and combinatorial immunotherapy approaches which include IC inhibiting antibodies and miR-34a, for EBV-associated cancers. Citation Format: Anastasiadou Eleni, Stroopinsky Dina, Stella Alimperti, Alan Jiao, Athalia Pyzer, Claudia Cipitelli, Martina Severa, Christopher S. Chen, Stefania Uccini, David Avigan, Alberto Faggioni, Pankaj Trivedi, Frank J. Slack. Epstein-Barr virus encoded EBNA2 alters immune checkpoint PD-L1 expression by downregulating miR-34a in B cell lymphomas [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A16.
癌细胞通过改变免疫检查点(IC)蛋白来破坏宿主的免疫监视。事实上,eb病毒(EBV)阳性的霍奇金淋巴瘤和胃腺癌具有较高的程序性细胞死亡配体PD-L1表达。然而,EBV如何在其首选宿主(B淋巴细胞和淋巴瘤)的背景下改变ic尚不清楚。在这里,我们使用伯基特淋巴瘤(BL),弥漫性大B细胞淋巴瘤(DLBCL)及其EBV感染或EBNA2转染衍生物来解决这个问题。EBV潜伏期III细胞表达高水平的PD-L1。在DLBCL模型中,我们发现EBNA2而不是LMP1足以诱导PD-L1。在雌激素诱导的携带EBNA2的B淋巴瘤细胞中证实了PD-L1的上调。来自DLBCL患者的临床样本显示,EBV感染的潜伏期III病例表达高水平的PD-L1。靶向肿瘤抑制因子miR-34a的PD-L1在EBNA2转染的细胞中下调。在表达DLBCL的EBNA2中,miR-34a重组可降低PD-L1的表达,并增加其在2D混合淋巴细胞反应(MLR)和3D微流控芯片MLR中的免疫原性。鉴于PD-L1抑制在癌症免疫治疗和miR-34a失调中的重要性,我们的研究结果可能对ebv相关癌症的诊断和组合免疫治疗方法(包括IC抑制抗体和miR-34a)具有重要意义。引文格式:Anastasiadou Eleni, Stroopinsky Dina, Stella Alimperti, Alan Jiao, Athalia Pyzer, Claudia Cipitelli, Martina Severa, Christopher S. Chen, Stefania Uccini, David Avigan, Alberto Faggioni, Pankaj Trivedi, Frank J. Slackeb病毒编码的EBNA2通过下调miR-34a改变B细胞淋巴瘤免疫检查点PD-L1的表达[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫学杂志,2018;6(9增刊):摘要nr A16。
{"title":"Abstract A16: Epstein-Barr virus encoded EBNA2 alters immune checkpoint PD-L1 expression by downregulating miR-34a in B cell lymphomas","authors":"Anastasiadou Eleni, Stroopinsky Dina, Stella Alimperti, Alan L Jiao, A. Pyzer, Claudia Cipitelli, M. Severa, Christopher S. Chen, S. Uccini, D. Avigan, A. Faggioni, P. Trivedi, F. Slack","doi":"10.1158/2326-6074.TUMIMM17-A16","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A16","url":null,"abstract":"Cancer cells subvert host immune surveillance by altering immune checkpoint (IC) proteins. Indeed, Epstein-Barr virus (EBV) positive Hodgkin’s lymphoma and gastric adenocarcinomas have higher Programmed Cell Death Ligand, PD-L1, expression. However, how EBV alters ICs in the context of its preferred host, the B lymphocyte and lymphomas, is unknown. Here, we used Burkitt lymphoma (BL), diffuse large B cell lymphomas (DLBCL) and their EBV infected or EBNA2 transfected derivatives to address this question. EBV latency III cells expressed high levels of PD-L1. In a DLBCL model, we found that EBNA2 but not LMP1 is sufficient to induce PD-L1. The upregulation of PD-L1 was confirmed in estrogen inducible EBNA2 carrying B lymphoma cells. Clinical samples from DLBCL patients showed that EBV infected, latency III cases expressed high levels of PD-L1. The PD-L1 targeting oncosuppressor miR-34a was downregulated in EBNA2 transfected cells. miR-34a reconstitution in EBNA2 expressing DLBCL reduced PD-L1 expression and increased their immunogenicity in 2D mixed lymphocyte reactions (MLR) and 3D microfluidic chip based MLRs. Given the importance of PD-L1 inhibition in immunotherapy and miR-34a dysregulation in cancers, our findings may have important implications for diagnosis and combinatorial immunotherapy approaches which include IC inhibiting antibodies and miR-34a, for EBV-associated cancers. Citation Format: Anastasiadou Eleni, Stroopinsky Dina, Stella Alimperti, Alan Jiao, Athalia Pyzer, Claudia Cipitelli, Martina Severa, Christopher S. Chen, Stefania Uccini, David Avigan, Alberto Faggioni, Pankaj Trivedi, Frank J. Slack. Epstein-Barr virus encoded EBNA2 alters immune checkpoint PD-L1 expression by downregulating miR-34a in B cell lymphomas [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A16.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"80334948","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 A12: Identification of resistance to immune checkpoint blockade A12:免疫检查点阻断的耐药性鉴定
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A12
Shengqing Gu, Ziyi Li, Xia Bu, Xiaofang Xing, G. Freeman, Myles A. Brown, Xiaole Shirley Liu
Despite its enormous success in treating several types of cancer, including melanoma, lung cancer, renal cancer, bladder cancer, and Hodgkin’s lymphoma, immunotherapy still only induces responses in a subset of patients. Understanding the response and resistance mechanisms are still open questions. Based on the reported heterogeneous response to targeted therapies in multiple cell lines, we hypothesize that significant heterogeneity might also exist regarding the response to immune checkpoint blockade (ICB), which may manifest the mechanisms of response/resistance to ICB. To this end, we applied the ClonTracer barcoding system to assess the heterogeneity of response to anti-PD1 and anti-CTLA4 in CT26 colorectal cancer model. Surprisingly, whereas significant heterogeneity exists in the cell line’s ability to initiate cancer in vivo, ICB treatment did not lead to dramatic clonal enrichment compared to control treatment. Therefore, the tumor microenvironment and/or the epigenetic programs in cancer cells may dictate the response to ICB. Note: This abstract was not presented at the conference. Citation Format: Shengqing Stanley Gu Gu, Ziyi Li, Xia Bu, Xiaofang Xing, Gordon Freeman, Myles Brown, Xiaole Shirley Liu. Identification of resistance to immune checkpoint blockade [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A12.
尽管免疫疗法在治疗包括黑色素瘤、肺癌、肾癌、膀胱癌和霍奇金淋巴瘤在内的几种癌症方面取得了巨大的成功,但它仍然只能在一小部分患者中引起反应。了解反应和抵抗机制仍然是一个悬而未决的问题。基于已报道的多种细胞系对靶向治疗的异质性反应,我们假设对免疫检查点阻断(ICB)的反应也可能存在显著的异质性,这可能表明对ICB的反应/耐药机制。为此,我们应用ClonTracer条形码系统评估CT26结直肠癌模型中抗pd1和抗ctla4反应的异质性。令人惊讶的是,尽管细胞系在体内启动癌症的能力存在显著的异质性,但与对照治疗相比,ICB治疗并未导致显著的克隆富集。因此,肿瘤微环境和/或癌细胞的表观遗传程序可能决定了对ICB的反应。注:本摘要未在会议上发表。引文格式:顾圣青、李子怡、布霞、邢晓芳、Gordon Freeman、Myles Brown、Xiaole Shirley Liu。免疫检查点阻断抵抗的鉴定[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫学杂志,2018;6(9增刊):摘要nr - A12。
{"title":"Abstract A12: Identification of resistance to immune checkpoint blockade","authors":"Shengqing Gu, Ziyi Li, Xia Bu, Xiaofang Xing, G. Freeman, Myles A. Brown, Xiaole Shirley Liu","doi":"10.1158/2326-6074.TUMIMM17-A12","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A12","url":null,"abstract":"Despite its enormous success in treating several types of cancer, including melanoma, lung cancer, renal cancer, bladder cancer, and Hodgkin’s lymphoma, immunotherapy still only induces responses in a subset of patients. Understanding the response and resistance mechanisms are still open questions. Based on the reported heterogeneous response to targeted therapies in multiple cell lines, we hypothesize that significant heterogeneity might also exist regarding the response to immune checkpoint blockade (ICB), which may manifest the mechanisms of response/resistance to ICB. To this end, we applied the ClonTracer barcoding system to assess the heterogeneity of response to anti-PD1 and anti-CTLA4 in CT26 colorectal cancer model. Surprisingly, whereas significant heterogeneity exists in the cell line’s ability to initiate cancer in vivo, ICB treatment did not lead to dramatic clonal enrichment compared to control treatment. Therefore, the tumor microenvironment and/or the epigenetic programs in cancer cells may dictate the response to ICB. Note: This abstract was not presented at the conference. Citation Format: Shengqing Stanley Gu Gu, Ziyi Li, Xia Bu, Xiaofang Xing, Gordon Freeman, Myles Brown, Xiaole Shirley Liu. Identification of resistance to immune checkpoint blockade [abstract]. In: Proceedings of the AACR Special Conference on Tumor Immunology and Immunotherapy; 2017 Oct 1-4; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2018;6(9 Suppl):Abstract nr A12.","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87200160","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 A07: Changes in immune profiles of osteosarcoma dogs receiving a GD3-based vaccine concurrently with carboplatin chemotherapy and surgery 摘要:骨肉瘤犬接受gd3疫苗联合卡铂化疗和手术后免疫谱的变化
Pub Date : 2018-09-01 DOI: 10.1158/2326-6074.TUMIMM17-A07
B. Sahay, S. Hutchison, Matt Cascio, A. Lejeune, C. Souza, A. Szivek, K. Shiomitsu, K. Harding, Stacey A Fox-Alvarez, Mia Livaccarri, L. Powers, R. Milner
Introduction: The disialyl gangliosides GD2/GD3 have been implicated in the enhancement of malignancy in a number of human and animal cancers and as a tumor antigen target for immunotherapy. In a recent abstract presented at an AACR Conference we reported on the coexpression of GD2/GD3 in four canine osteosarcoma (OSA) cell lines (1). In a prospective IACUC approved clinical trial we vaccinated dogs with a GD3-based vaccine with naturally occurring OSA receiving surgery and carboplatin chemotherapy to investigate the expression profiles of immune modulating cells overtime. Methods: Dogs will be entered into the study only if they meet the following inclusion criteria: have a confirmed diagnosis of OSA and no other life-threatening diseases. The study will accrue 40 cases; 20 will receive the vaccine plus standard of care and 20 dogs will receive only the standard of care (amputation and intent to treat with 6 doses of carboplatin). On admission blood will be collected according to the protocol for monitoring of the immune response. The dogs will then be vaccinated according to a predetermined protocol during chemotherapy and staged. The immune response profile of the vaccine group will be compared to dogs receiving standard of care alone and normal dogs. Flow cytometric platforms were developed to monitor changes in immune cells (CD5, CD21, CD4, CD8, CD14, CD11b, MHCII, and Foxp3). In addition, IHC arrays and RNA Scope will be developed for checkpoints of immunity PD1, PDL-1 and expression of intratumoral immune cells. Results: Currently twenty dogs with osteosarcoma have enrolled into the study and have received standard of care and vaccination. Complete flow cytometric immune profiles are available for nine dogs with osteosarcoma. On admission, all dogs with OSA showed elevated cell counts of Treg (FoxP3+/CD4+) cells, Monocytic (m-) and Granulocytic (g-) myeloid derived suppressor cells (MDSCs) when compared to normal dogs (all dogs had normal CBCs). All m-MDCSs and g-MDSCs and Treg cells decreased significantly after the first dose of chemotherapy. Serial sampling over weeks showed sustained inhibition even after chemotherapy was completed (18 weeks). Two OSA cases which relapsed with metastasis to the lungs showed significant increases in Treg cells at the time of restaging. Complete necropsy post-therapy in three dogs showed changes in metastatic profiles; 2/3 showed no metastatic disease to the lungs, but metastases occurred to bone and kidneys. Discussion: MDSCs series of cells and Treg cells are increased in OSA compared to normal dogs and may be a factor in maintaining a welcoming tumor microenvironment and resistance to immunotherapy. MDSCs are reported to be elevated in other cancers, but not in naturally occurring OSA. Furthermore, canine OSA cell lines show increased expression of CCL2 and COX2 (data not shown). Recruitment of MDSCs to the cancer micro-tumor environment are thought to be mediated by the chemokine CCL2 and COX2. Abrog
简介:双醚神经节苷脂GD2/GD3在许多人类和动物癌症中与恶性肿瘤的增强有关,并作为免疫治疗的肿瘤抗原靶点。最近在AACR会议上发表的一篇摘要中,我们报道了GD2/GD3在四种犬骨肉瘤(OSA)细胞系中的共表达(1)。在一项IACUC批准的前瞻性临床试验中,我们给患有自然发生OSA的狗接种了基于GD3的疫苗,接受手术和卡铂化疗,以研究免疫调节细胞的表达谱。方法:符合以下入选标准的犬只将被纳入研究:确诊为OSA且无其他危及生命的疾病。该研究将收集40例病例;20只狗将接受疫苗加标准护理,20只狗将接受标准护理(截肢和打算用6剂卡铂治疗)。入院时将根据免疫反应监测方案采血。然后,这些狗将在化疗期间按照预先确定的方案接种疫苗,并分阶段接种。接种疫苗组的免疫反应将与单独接受标准护理的狗和正常狗进行比较。开发了流式细胞仪平台来监测免疫细胞(CD5、CD21、CD4、CD8、CD14、CD11b、MHCII和Foxp3)的变化。此外,将开发免疫组化阵列和RNA Scope用于免疫PD1、PDL-1和肿瘤内免疫细胞表达的检查点。结果:目前已有20只患有骨肉瘤的狗参加了这项研究,并接受了标准的护理和疫苗接种。完整的流式细胞术免疫图谱可用于9只骨肉瘤犬。入院时,与正常犬相比,所有OSA犬的Treg (FoxP3+/CD4+)细胞、单核细胞(m-)和粒细胞(g-)骨髓源性抑制细胞(MDSCs)计数均升高(所有犬的CBCs正常)。所有m- mdscs、g-MDSCs和Treg细胞在第一次化疗后均显著减少。持续数周的连续取样显示,即使在化疗完成后(18周),也能持续抑制。2例复发并肺转移的OSA患者在重新分期时Treg细胞明显增加。治疗后的三只狗的完全尸检显示转移谱的变化;2/3未出现肺转移,但发生骨和肾转移。讨论:与正常犬相比,OSA患者的MDSCs系列细胞和Treg细胞增加,这可能是维持肿瘤微环境和免疫治疗抵抗的一个因素。据报道,MDSCs在其他癌症中升高,但在自然发生的OSA中没有升高。此外,犬OSA细胞系显示CCL2和COX2的表达增加(数据未显示)。MDSCs向癌症微肿瘤环境的募集被认为是由趋化因子CCL2和COX2介导的。化疗和可能的免疫治疗可以消除这些途径,提高总生存率。早期尸检数据似乎支持接受标准护理和基于gd3的疫苗的狗的转移谱衰减。结论:该研究的预期结果将用于根据免疫特征的变化调整疫苗方案。目前的数据还不够成熟,不足以评估生存率。致谢:该研究由美国养犬俱乐部健康基金会和UF CVM资助。参考:1。【摘要】:神经节苷脂GD3和GD2在犬和人骨肉瘤细胞中的差异表达:免疫治疗靶点。癌症免疫杂志2015年10月1日;3(10增刊):A29。引文格式:Bikash Sahay, Shana Hutchison, Matt Cascio, Amandine Lejeune, Carlos Souza, Anna Szivek, Keijiro Shiomitsu, Kayla Harding, Stacey Fox-Alvarez, Mia Livaccarri, Lindsay Powers, Rowan James Milner。骨肉瘤犬接受gd3疫苗联合卡铂化疗和手术后免疫谱的变化[摘要]。摘自:AACR肿瘤免疫学和免疫治疗特别会议论文集;2017年10月1-4日;波士顿,MA。费城(PA): AACR;癌症免疫杂志,2018;6(9增刊):摘要nr A07。
{"title":"Abstract A07: Changes in immune profiles of osteosarcoma dogs receiving a GD3-based vaccine concurrently with carboplatin chemotherapy and surgery","authors":"B. Sahay, S. Hutchison, Matt Cascio, A. Lejeune, C. Souza, A. Szivek, K. Shiomitsu, K. Harding, Stacey A Fox-Alvarez, Mia Livaccarri, L. Powers, R. Milner","doi":"10.1158/2326-6074.TUMIMM17-A07","DOIUrl":"https://doi.org/10.1158/2326-6074.TUMIMM17-A07","url":null,"abstract":"Introduction: The disialyl gangliosides GD2/GD3 have been implicated in the enhancement of malignancy in a number of human and animal cancers and as a tumor antigen target for immunotherapy. In a recent abstract presented at an AACR Conference we reported on the coexpression of GD2/GD3 in four canine osteosarcoma (OSA) cell lines (1). In a prospective IACUC approved clinical trial we vaccinated dogs with a GD3-based vaccine with naturally occurring OSA receiving surgery and carboplatin chemotherapy to investigate the expression profiles of immune modulating cells overtime. Methods: Dogs will be entered into the study only if they meet the following inclusion criteria: have a confirmed diagnosis of OSA and no other life-threatening diseases. The study will accrue 40 cases; 20 will receive the vaccine plus standard of care and 20 dogs will receive only the standard of care (amputation and intent to treat with 6 doses of carboplatin). On admission blood will be collected according to the protocol for monitoring of the immune response. The dogs will then be vaccinated according to a predetermined protocol during chemotherapy and staged. The immune response profile of the vaccine group will be compared to dogs receiving standard of care alone and normal dogs. Flow cytometric platforms were developed to monitor changes in immune cells (CD5, CD21, CD4, CD8, CD14, CD11b, MHCII, and Foxp3). In addition, IHC arrays and RNA Scope will be developed for checkpoints of immunity PD1, PDL-1 and expression of intratumoral immune cells. Results: Currently twenty dogs with osteosarcoma have enrolled into the study and have received standard of care and vaccination. Complete flow cytometric immune profiles are available for nine dogs with osteosarcoma. On admission, all dogs with OSA showed elevated cell counts of Treg (FoxP3+/CD4+) cells, Monocytic (m-) and Granulocytic (g-) myeloid derived suppressor cells (MDSCs) when compared to normal dogs (all dogs had normal CBCs). All m-MDCSs and g-MDSCs and Treg cells decreased significantly after the first dose of chemotherapy. Serial sampling over weeks showed sustained inhibition even after chemotherapy was completed (18 weeks). Two OSA cases which relapsed with metastasis to the lungs showed significant increases in Treg cells at the time of restaging. Complete necropsy post-therapy in three dogs showed changes in metastatic profiles; 2/3 showed no metastatic disease to the lungs, but metastases occurred to bone and kidneys. Discussion: MDSCs series of cells and Treg cells are increased in OSA compared to normal dogs and may be a factor in maintaining a welcoming tumor microenvironment and resistance to immunotherapy. MDSCs are reported to be elevated in other cancers, but not in naturally occurring OSA. Furthermore, canine OSA cell lines show increased expression of CCL2 and COX2 (data not shown). Recruitment of MDSCs to the cancer micro-tumor environment are thought to be mediated by the chemokine CCL2 and COX2. Abrog","PeriodicalId":9948,"journal":{"name":"Checkpoints and Immunomodulation","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2018-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"87761328","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
期刊
Checkpoints and Immunomodulation
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