首页 > 最新文献

Genetically Engineered T-cells最新文献

英文 中文
Abstract A025: Eradication of neuroblastoma by T-cells redirected with an optimized GD2-specific chimeric antigen receptor and IL-15 利用优化的gd2特异性嵌合抗原受体和IL-15重定向t细胞根除神经母细胞瘤
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A025
Yuhui Chen, Chuang Sun, L. Metelitsa, G. Dotti, B. Savoldo
The treatment with CD19-specific chimeric antigen receptor T-cells (CAR-Ts) has shown remarkable antitumor activity in patients with B-cell malignancies. However, the clinical benefits of CAR-Ts in solid tumors remain unclear. A compelling concern is that in patients with solid tumors CAR-Ts do not immediately encounter their cognate antigen in the circulation and thus lack the appropriate costimulatory signals necessary for full activation. We here sought to explore if expressing IL-15 in CAR-Ts would provide them with sufficient sustained survival until they engage the cognate antigen. Using the GD2-specific CAR and neuroblastoma (NB) as a tumor model, we explored the benefits of incorporating the IL-15 cytokine (and the iCaspase9 suicide gene for safety) within the CAR molecule. CAR-Ts from 12 healthy donors were transduced with an optimized GD2.CAR (encoding the CD28 endodomain) without (GD2-Ts) or with the IL15 (GD2.15.iC9-Ts) and expanded ex vivo with IL-7/IL-15 for 14 days. CAR transduction (82% ± 8% and 82% ± 12%, respectively) and ex vivo antitumor activity in 4 days co-culture at different E:T ratios were comparable. However, upon repetitive stimulation with GD2+ NB tumors (CHLA-255 and LAN-1), only GD2.15.iC9-Ts showed significantly superior expansion and antitumor activity (p Citation Format: Yuhui Chen, Chuang Sun, Leonid Metelitsa, Gianpietro Dotti, Barbara Savoldo. Eradication of neuroblastoma by T-cells redirected with an optimized GD2-specific chimeric antigen receptor and IL-15 [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 A025.
cd19特异性嵌合抗原受体t细胞(CAR-Ts)治疗在b细胞恶性肿瘤患者中显示出显著的抗肿瘤活性。然而,car - t治疗实体瘤的临床益处尚不清楚。一个令人关注的问题是,在患有实体肿瘤的患者中,car - t不能立即在循环中遇到它们的同源抗原,因此缺乏充分激活所需的适当共刺激信号。我们试图探索如果表达IL-15 CAR-Ts将为他们提供足够的持续生存,直到它们同源抗原接触。使用gd2特异性CAR和神经母细胞瘤(NB)作为肿瘤模型,我们探索了在CAR分子中加入IL-15细胞因子(以及iCaspase9自杀基因)的益处。来自12名健康供体的car - t用优化的GD2转导。CAR(编码CD28内结构域)不含GD2-Ts或含有IL-15 (GD2.15.iC9-Ts),并在体外用IL-7/IL-15扩增14天。CAR转导率(分别为82%±8%和82%±12%)和体外抗肿瘤活性在不同E:T比下共培养4天具有可比性。然而,在GD2+ NB肿瘤(CHLA-255和LAN-1)的重复刺激下,只有GD2.15。iC9-Ts具有显著的扩展和抗肿瘤活性(p引文格式:陈宇晖,孙创,Leonid Metelitsa, Gianpietro Dotti, Barbara Savoldo)。用优化的gd2特异性嵌合抗原受体和IL-15重定向t细胞根除神经母细胞瘤[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 A025。
{"title":"Abstract A025: Eradication of neuroblastoma by T-cells redirected with an optimized GD2-specific chimeric antigen receptor and IL-15","authors":"Yuhui Chen, Chuang Sun, L. Metelitsa, G. Dotti, B. Savoldo","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A025","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A025","url":null,"abstract":"The treatment with CD19-specific chimeric antigen receptor T-cells (CAR-Ts) has shown remarkable antitumor activity in patients with B-cell malignancies. However, the clinical benefits of CAR-Ts in solid tumors remain unclear. A compelling concern is that in patients with solid tumors CAR-Ts do not immediately encounter their cognate antigen in the circulation and thus lack the appropriate costimulatory signals necessary for full activation. We here sought to explore if expressing IL-15 in CAR-Ts would provide them with sufficient sustained survival until they engage the cognate antigen. Using the GD2-specific CAR and neuroblastoma (NB) as a tumor model, we explored the benefits of incorporating the IL-15 cytokine (and the iCaspase9 suicide gene for safety) within the CAR molecule. CAR-Ts from 12 healthy donors were transduced with an optimized GD2.CAR (encoding the CD28 endodomain) without (GD2-Ts) or with the IL15 (GD2.15.iC9-Ts) and expanded ex vivo with IL-7/IL-15 for 14 days. CAR transduction (82% ± 8% and 82% ± 12%, respectively) and ex vivo antitumor activity in 4 days co-culture at different E:T ratios were comparable. However, upon repetitive stimulation with GD2+ NB tumors (CHLA-255 and LAN-1), only GD2.15.iC9-Ts showed significantly superior expansion and antitumor activity (p Citation Format: Yuhui Chen, Chuang Sun, Leonid Metelitsa, Gianpietro Dotti, Barbara Savoldo. Eradication of neuroblastoma by T-cells redirected with an optimized GD2-specific chimeric antigen receptor and IL-15 [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 A025.","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"135 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"127499019","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: 5
Abstract A038: Effective rerouting of NK cell cytotoxicity against B-cell malignancies upon TCR gene transfer 通过TCR基因转移NK细胞对b细胞恶性肿瘤细胞毒性的有效改变
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A038
Laura T Morton, A. Wouters, D. Remst, R. Hagedoorn, M. Loenen, R. Boer, J. H. Falkenberg, M. Heemskerk
T-cell receptor (TCR) gene transfer involves ex-vivo introduction of a tumour-reactive TCR into patient-derived CD8 T-cells enabling specific-targeting of tumour cells. Competition for expression with CD3 from the endogenous TCR and the potential for TCR mixed- dimer formation necessitate optimisation of cellular therapeutics with sustained potency and increased safety. NK-cells (CD3-CD56+) are potent short-lived effector cells that lyse abnormal or stressed cells independent of antigen. Efficacy and safety of adoptive NK therapy has been demonstrated in the treatment of hematologic malignancies in both the autologous and allogeneic setting. Here,we aimed to exploit NK-cell cytotoxicity and redirect it toward antigen-specific recognition of tumors without the limitation of TCR mixed- dimer formation and competition for CD3.Firstly, peripheral blood derived NK-cells were expanded and retrovirally transduced to express BOB1-specific TCR, restricted to HLA- B*07:02, in combination with CD3 alongside CD8 co-receptor.BOB1 is a B-cell restricted transcription factor, important for B-cell survival. Purified BOB1-TCR expressing NK-cells demonstrated antigen-specific binding of BOB1-specific pMHC-tetramer and proliferated upon co-culture with HLA-B*07:02 positive B-lymphoblastic cell lines (B-LCL) but not with HLA-B*07:02 negative B-LCL. Furthermore, BOB1-TCR expressing NK-cells demonstrated in vitro cytotoxicity against HLA-B*07:02 positive B-LCL, multiple myeloma and B-ALL cell lines. Conversely, these tumor cell lines remained resistant to NK-cell mediated lysis when co-cultured with mock transduced NK-cells. Finally, NK sensitive cell line K562 was comparably lysed by both BOB1-TCR or mock transduced NK-cells demonstrating retained NK-cell mediated activity.These data demonstrated that NK-cell cytotoxicity can be redirected toward antigen-specific recognition of tumors and is TCR-dependent. Retention of NK-cell function in genetically modified cells allows for a double-hit therapeutic approach that can offer advantages over current cellular approaches. Citation Format: Laura T. Morton, Anne K. Wouters, Dennis F. Remst, Renate S. Hagedoorn, Marleen M. Van Loenen, Renate de Boer, J. H.F. Falkenberg, Mirjam H.M. Heemskerk. Effective rerouting of NK cell cytotoxicity against B-cell malignancies upon TCR gene transfer [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 A038.
t细胞受体(TCR)基因转移涉及将肿瘤反应性TCR体外导入患者来源的CD8 t细胞中,从而实现肿瘤细胞的特异性靶向。内源性TCR与CD3的表达竞争以及TCR混合二聚体形成的潜力需要优化具有持续效力和增加安全性的细胞治疗方法。nk细胞(CD3-CD56+)是一种有效的短寿命效应细胞,可以独立于抗原溶解异常或应激细胞。过继NK疗法的有效性和安全性已经在自体和异体血液恶性肿瘤的治疗中得到证实。在这里,我们的目标是利用nk细胞的细胞毒性,并将其定向到肿瘤抗原特异性识别,而不受TCR混合二聚体形成和CD3竞争的限制。首先,外周血源性nk细胞扩增并经逆转录转导表达bob1特异性TCR,该TCR仅限于HLA- B*07:02,与CD3和CD8共受体结合。BOB1是一种b细胞限制性转录因子,对b细胞存活至关重要。纯化后表达nk细胞的BOB1-TCR能与bob1特异性pmhc -四聚体结合,并能与HLA-B*07:02阳性的b淋巴母细胞(B-LCL)共培养增殖,而与HLA-B*07:02阴性的B-LCL共培养不增殖。此外,表达BOB1-TCR的nk细胞对HLA-B*07:02阳性的B-LCL、多发性骨髓瘤和B-ALL细胞系显示出体外细胞毒性。相反,当与模拟转导的nk细胞共培养时,这些肿瘤细胞系仍然对nk细胞介导的裂解具有抗性。最后,NK敏感细胞系K562被BOB1-TCR或模拟转导的NK细胞裂解,显示保留NK细胞介导的活性。这些数据表明,nk细胞的细胞毒性可以重定向到抗原特异性识别肿瘤,并且依赖于tcr。在基因修饰的细胞中保留nk细胞功能允许双重打击治疗方法,可以提供优于当前细胞方法的优势。引文格式:Laura T. Morton, Anne K. Wouters, Dennis F. Remst, Renate S. Hagedoorn, Marleen M. Van Loenen, Renate de Boer, j.h.f. Falkenberg, Mirjam H.M. Heemskerk。TCR基因转移后NK细胞对b细胞恶性肿瘤细胞毒性的有效重定向[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 A038。
{"title":"Abstract A038: Effective rerouting of NK cell cytotoxicity against B-cell malignancies upon TCR gene transfer","authors":"Laura T Morton, A. Wouters, D. Remst, R. Hagedoorn, M. Loenen, R. Boer, J. H. Falkenberg, M. Heemskerk","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A038","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A038","url":null,"abstract":"T-cell receptor (TCR) gene transfer involves ex-vivo introduction of a tumour-reactive TCR into patient-derived CD8 T-cells enabling specific-targeting of tumour cells. Competition for expression with CD3 from the endogenous TCR and the potential for TCR mixed- dimer formation necessitate optimisation of cellular therapeutics with sustained potency and increased safety. NK-cells (CD3-CD56+) are potent short-lived effector cells that lyse abnormal or stressed cells independent of antigen. Efficacy and safety of adoptive NK therapy has been demonstrated in the treatment of hematologic malignancies in both the autologous and allogeneic setting. Here,we aimed to exploit NK-cell cytotoxicity and redirect it toward antigen-specific recognition of tumors without the limitation of TCR mixed- dimer formation and competition for CD3.Firstly, peripheral blood derived NK-cells were expanded and retrovirally transduced to express BOB1-specific TCR, restricted to HLA- B*07:02, in combination with CD3 alongside CD8 co-receptor.BOB1 is a B-cell restricted transcription factor, important for B-cell survival. Purified BOB1-TCR expressing NK-cells demonstrated antigen-specific binding of BOB1-specific pMHC-tetramer and proliferated upon co-culture with HLA-B*07:02 positive B-lymphoblastic cell lines (B-LCL) but not with HLA-B*07:02 negative B-LCL. Furthermore, BOB1-TCR expressing NK-cells demonstrated in vitro cytotoxicity against HLA-B*07:02 positive B-LCL, multiple myeloma and B-ALL cell lines. Conversely, these tumor cell lines remained resistant to NK-cell mediated lysis when co-cultured with mock transduced NK-cells. Finally, NK sensitive cell line K562 was comparably lysed by both BOB1-TCR or mock transduced NK-cells demonstrating retained NK-cell mediated activity.These data demonstrated that NK-cell cytotoxicity can be redirected toward antigen-specific recognition of tumors and is TCR-dependent. Retention of NK-cell function in genetically modified cells allows for a double-hit therapeutic approach that can offer advantages over current cellular approaches. Citation Format: Laura T. Morton, Anne K. Wouters, Dennis F. Remst, Renate S. Hagedoorn, Marleen M. Van Loenen, Renate de Boer, J. H.F. Falkenberg, Mirjam H.M. Heemskerk. Effective rerouting of NK cell cytotoxicity against B-cell malignancies upon TCR gene transfer [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 A038.","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"22 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"122068016","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 A024: Identifying a T-cell receptor for immunotherapy against a leukemia-associated self-antigen in an allogeneic setting 摘要/ Abstract A024:在异基因环境中鉴定用于免疫治疗白血病相关自身抗原的t细胞受体
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A024
Maxi-Lu Böschen, Weiwen Yang, E. Strønen, J. Olweus
Our aim was to identify a T-cell receptor (TCR) that recognizes a leukemia-associated self-antigen that is highly expressed in acute myeloid leukemia and that is restricted by HLA-A2 (expressed in approximately 50% of Caucasians) for future adoptive T-cell therapy. Tumor-associated self-antigens (self-TAA) make attractive targets for adoptive T-cell therapy in cancer. However, autologous T-cells are tolerant to self-TAA expressed on self-HLA if antigen expression is sufficiently high. In contrast, T-cells are not tolerant to self-antigens presented on non-self HLA. By identifying alloreactive T-cells reactive to self-peptide in complex with foreign HLA and subsequently cloning their TCR, it is possible to redirect patient T-cells against self-TAA. The advantage of targeting self-antigens is the potential application of TCR-engineered T-cells in all cancer patients with a certain cancer type and expressing the restricting HLA molecule. We identified a protein that is selectively expressed in normal myeloid cells and highly expressed by leukemic cells in patients with acute myeloid leukemia (AML). HLA-A2 negative CD8 T-cells from healthy donors were stimulated with HLA-A2 positive dendritic cells that were expressing the full-length target antigen. CD8 T-cells recognizing peptides from the target antigen in complex with HLA-A2 were subsequently identified and sorted using fluorescently labeled HLA-A2-peptide-multimers. T-cell clones obtained from sorted multimer-positive T-cells responded to HLA-A2 positive patient leukemia cells and cell lines expressing the antigen, whereas no response was seen to HLA-A2 positive antigen negative targeT-cells, unless loaded with the relevant peptide. Responses were measured as secretion of interferon gamma, expression of the T-cell activation marker CD137 or expression of the degranulation marker CD107. The TCR sequences were identified, and we next evaluated specificity and functionality of TCR-engineered cells. Indeed, TCR-transduced healthy donor T-cells were able to selectively kill HLA-A2 positive AML patient-derived leukemic cells as well as antigen positive cell lines in vitro. We also performed a comprehensive peptide scan to determine potential cross reactivity of the obtained TCR. For this scan, the amino acids (AA) in each position of the target peptide-epitope were individually replaced by every other natural amino acid (19 AA for each position x 9 postions = 171 peptides). In addition, potential responses to other variants of the 9mer target peptide based on the natural antigen protein sequence were evaluated (8mer, 10mers, 11mers and 12mers). Based on the results obtained with TCR-transduced T-cells (interferon gamma ELISA), candidate peptides naturally occurring in the proteome to which our TCR could cross-react were not identified. These data warrant further evaluation of the TCR for in vivo efficacy and specificity. Citation Format: Maxi-Lu Boschen, Weiwen Yang, Erlend Stronen, Johanna Olweus. Id
我们的目的是鉴定一种t细胞受体(TCR),它可以识别白血病相关的自身抗原,这种抗原在急性髓性白血病中高度表达,并且受HLA-A2(在大约50%的高加索人中表达)的限制,用于未来的过继t细胞治疗。肿瘤相关的自身抗原(self-TAA)成为过继t细胞治疗癌症的有吸引力的靶点。然而,如果抗原表达足够高,自体t细胞对自身hla上表达的自身taa具有耐受性。相反,t细胞不能耐受非自体HLA上的自体抗原。通过鉴定与外源HLA复合物中对自身肽反应的同种异体反应性t细胞,并随后克隆其TCR,有可能使患者t细胞重定向对抗自身taa。靶向自身抗原的优势在于tcr工程t细胞在所有具有某种癌症类型并表达限制性HLA分子的癌症患者中的潜在应用。我们发现了一种在正常髓细胞中选择性表达的蛋白,在急性髓性白血病(AML)患者的白血病细胞中高度表达。来自健康供体的HLA-A2阴性CD8 t细胞被表达全长靶抗原的HLA-A2阳性树突状细胞刺激。随后使用荧光标记的HLA-A2多肽多聚体对识别目标抗原多肽的CD8 t细胞进行鉴定和分类。从分选的多阳性t细胞中获得的t细胞克隆对HLA-A2阳性患者白血病细胞和表达抗原的细胞系有反应,而对HLA-A2阳性抗原阴性靶细胞没有反应,除非加载相关肽。通过干扰素γ的分泌、t细胞活化标志物CD137的表达或脱颗粒标志物CD107的表达来测量应答。鉴定了TCR序列,然后我们评估了TCR工程细胞的特异性和功能。事实上,在体外,tcr转导的健康供体t细胞能够选择性地杀死HLA-A2阳性AML患者来源的白血病细胞以及抗原阳性细胞系。我们还进行了全面的肽扫描,以确定获得的TCR的潜在交叉反应性。在这次扫描中,靶肽表位每个位置的氨基酸(AA)被其他天然氨基酸单独替换(每个位置19个AA x 9个位置= 171个肽)。此外,基于天然抗原蛋白序列,评估了对9mer靶肽其他变体(8mer、10mers、11mers和12mers)的潜在应答。根据TCR转导的t细胞(干扰素γ ELISA)获得的结果,TCR可以交叉反应的蛋白质组中自然存在的候选肽未被鉴定出来。这些数据为进一步评估TCR的体内疗效和特异性提供了依据。引用格式:Maxi-Lu Boschen, Weiwen Yang, Erlend Stronen, Johanna Olweus。在异基因环境中鉴定用于免疫治疗白血病相关自身抗原的t细胞受体[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 A024。
{"title":"Abstract A024: Identifying a T-cell receptor for immunotherapy against a leukemia-associated self-antigen in an allogeneic setting","authors":"Maxi-Lu Böschen, Weiwen Yang, E. Strønen, J. Olweus","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A024","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A024","url":null,"abstract":"Our aim was to identify a T-cell receptor (TCR) that recognizes a leukemia-associated self-antigen that is highly expressed in acute myeloid leukemia and that is restricted by HLA-A2 (expressed in approximately 50% of Caucasians) for future adoptive T-cell therapy. Tumor-associated self-antigens (self-TAA) make attractive targets for adoptive T-cell therapy in cancer. However, autologous T-cells are tolerant to self-TAA expressed on self-HLA if antigen expression is sufficiently high. In contrast, T-cells are not tolerant to self-antigens presented on non-self HLA. By identifying alloreactive T-cells reactive to self-peptide in complex with foreign HLA and subsequently cloning their TCR, it is possible to redirect patient T-cells against self-TAA. The advantage of targeting self-antigens is the potential application of TCR-engineered T-cells in all cancer patients with a certain cancer type and expressing the restricting HLA molecule. We identified a protein that is selectively expressed in normal myeloid cells and highly expressed by leukemic cells in patients with acute myeloid leukemia (AML). HLA-A2 negative CD8 T-cells from healthy donors were stimulated with HLA-A2 positive dendritic cells that were expressing the full-length target antigen. CD8 T-cells recognizing peptides from the target antigen in complex with HLA-A2 were subsequently identified and sorted using fluorescently labeled HLA-A2-peptide-multimers. T-cell clones obtained from sorted multimer-positive T-cells responded to HLA-A2 positive patient leukemia cells and cell lines expressing the antigen, whereas no response was seen to HLA-A2 positive antigen negative targeT-cells, unless loaded with the relevant peptide. Responses were measured as secretion of interferon gamma, expression of the T-cell activation marker CD137 or expression of the degranulation marker CD107. The TCR sequences were identified, and we next evaluated specificity and functionality of TCR-engineered cells. Indeed, TCR-transduced healthy donor T-cells were able to selectively kill HLA-A2 positive AML patient-derived leukemic cells as well as antigen positive cell lines in vitro. We also performed a comprehensive peptide scan to determine potential cross reactivity of the obtained TCR. For this scan, the amino acids (AA) in each position of the target peptide-epitope were individually replaced by every other natural amino acid (19 AA for each position x 9 postions = 171 peptides). In addition, potential responses to other variants of the 9mer target peptide based on the natural antigen protein sequence were evaluated (8mer, 10mers, 11mers and 12mers). Based on the results obtained with TCR-transduced T-cells (interferon gamma ELISA), candidate peptides naturally occurring in the proteome to which our TCR could cross-react were not identified. These data warrant further evaluation of the TCR for in vivo efficacy and specificity. Citation Format: Maxi-Lu Boschen, Weiwen Yang, Erlend Stronen, Johanna Olweus. Id","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"4 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"121334397","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 A023: Highly enriched memory stem T-cell subsets (TSCM) expressing a novel CAR30 have enhanced antitumor effect in Hodgkin lymphoma A023:表达一种新的CAR30的高度富集的记忆干细胞t细胞亚群(TSCM)增强了霍奇金淋巴瘤的抗肿瘤作用
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A023
C. Álvarez-Fernández, L. Escribà-García, A. Caballero, J. Sierra, J. Briones
Background: Up to 20% of patients with advanced Hodgkin lymphoma (HL) are not cured with current chemo-immunotherapy treatments. Adoptive immunotherapy (AIT) with mature T-cells modified with a chimeric antigen receptor (CAR) holds promise for the treatment of various types of B cell lymphoma. The CD30 antigen, expressed in virtually all HL tumor cells, is absent in most healthy tissues, thus representing an ideal target for AIT of HL. In contrast to CAR19 for B cell malignancies, efficacy of CAR30 T-cells for Hodgkin and other CD30 lymphomas remains modest. Thus, novel approaches are needed to significantly improve the clinical efficacy of this approach. To this end, we used a novel antigen binding domain of a CD30 mAb, unaffected by soluble CD30 protein, as part of a chimeric antigen receptor (CAR) coupled to the 4.1BB and ζ chain endodomains, and transduced memory stem T-cells (TSCM) in an effort to ensure efficient engraftment, prolonged persistence, and enhancement of in vivo antitumor activity. To evaluate the feasibility of ex vivo expansion and antitumor efficacy of genetically-modified TSCM with a novel CD30-specific CAR that recognizes a membrane-proximal epitope of the CD30 molecule for the treatment of HL. Methods: A second-generation CD30-41BBz-EGFRt CAR was developed using a scFv that recognizes a membrane-proximal epitope of the CD30 protein. TSCM cells were generated with IL-7, IL-15 and IL-21, and transduced on day 2 of culture with a third-generation lentiviral vector encoding the CD30-CAR. The HL derived cell line L540 was used as tumor model. Tumor-specific cytotoxicity and cytokines were analysed at 24 hours. The in vivo antitumor efficacy was tested in a NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mouse model of HL xenografted with 2,5x10^6 L450 cells. Results: After 10 days, TSCM represented the majority of T-cells in culture (50.38±5.47 % for CD4+ and 70.84 ± 3.36 % for CD8+ TSCM, respectively) with a very high expression of CD30-CAR (77 ± 3.64 % in CD4+ and 83.12 ± 4.25 % in CD8+ TSCM, respectively). Expression of CD30 protein in TSCM increased over the culture period, peaking at day 8 in both CD4+ (17.5 ± 4.17 %) and CD8+ TSCM (42.37 ± 5.75 %), respectively. Despite these high levels of CD30 expression, TSCM expanded ex vivo very efficiently (CD4+ CD30-CAR: 97.24 ± 26.2 fold expansion, and CD8+ CD30-CAR: 96.79 ± 22.84 fold expansion, respectively), although lower than TSCM transduced with a CD19-CAR (CD4+ CD19-CAR: 161.96±35.9 fold expansion, and CD8+ CD19-CAR: 163.25 ± 26.51, fold expansion, respectively). Bulky CD30-CAR TSCM mediated very efficient in vitro tumor cytotoxicity (tumor cell death at 5:1 ratio: 85.25% vs. 0% with untransduced TSCM), while control CD30- targeT-cells were not killed. In addition, we evaluated the antitumor efficacy of CD30-CAR TSCM in vivo. Upon adoptive transfer of 10x10^6 bulk (CD4+ and CD8+) TSCM into NSG mice engrafted with L540 (2,5x10^6 subcutaneously), complete eradication of HL was observ
背景:高达20%的晚期霍奇金淋巴瘤(HL)患者无法通过目前的化学免疫治疗治愈。用嵌合抗原受体修饰成熟t细胞的过继免疫疗法(AIT)有望治疗各种类型的B细胞淋巴瘤。CD30抗原在几乎所有HL肿瘤细胞中表达,在大多数健康组织中不存在,因此代表了HL AIT的理想靶点。与CAR19治疗B细胞恶性肿瘤相比,CAR30 t细胞治疗霍奇金淋巴瘤和其他CD30淋巴瘤的疗效仍然不高。因此,需要新的方法来显著提高该方法的临床疗效。为此,我们使用了一种新的CD30单抗抗原结合结构域,不受可溶性CD30蛋白的影响,作为嵌合抗原受体(CAR)的一部分,偶联到4.1BB和ζ链内结构域,并转导记忆干细胞t细胞(TSCM),以确保有效的植入,延长持久性,并增强体内抗肿瘤活性。利用一种新的CD30特异性CAR来评估基因修饰TSCM体外扩增的可行性和抗肿瘤效果,该CAR可识别CD30分子的膜近端表位,用于治疗HL。方法:使用识别CD30蛋白膜近端表位的scFv开发第二代CD30- 41bbz - egfrt CAR。用IL-7、IL-15和IL-21生成TSCM细胞,并在培养的第2天用编码CD30-CAR的第三代慢病毒载体进行转导。以HL衍生细胞系L540作为肿瘤模型。24小时检测肿瘤特异性细胞毒性和细胞因子。用NSG (NOD)检测其体内抗肿瘤效果。2,5 × 10^6 L450细胞移植HL小鼠模型。结果:10 d后,TSCM占培养t细胞的大多数(CD4+为50.38±5.47%,CD8+ TSCM为70.84±3.36%),CD30-CAR表达非常高(CD4+为77±3.64%,CD8+ TSCM为83.12±4.25%)。CD30蛋白在TSCM中的表达随着培养时间的增加而增加,CD4+(17.5±4.17%)和CD8+ TSCM(42.37±5.75%)的表达在第8天达到峰值。尽管CD30的表达水平很高,但TSCM的体外扩增效率非常高(CD4+ CD30- car: 97.24±26.2倍扩增,CD8+ CD30- car: 96.79±22.84倍扩增),尽管低于CD19-CAR转导的TSCM (CD4+ CD19-CAR: 161.96±35.9倍扩增,CD8+ CD19-CAR: 163.25±26.51倍扩增)。庞大的CD30- car TSCM介导了非常有效的体外肿瘤细胞毒性(肿瘤细胞死亡率为5:1:85.25% vs.未转导的TSCM为0%),而对照CD30-靶细胞未被杀死。此外,我们在体内评估了CD30-CAR - TSCM的抗肿瘤功效。将10 × 10^6体积(CD4+和CD8+) TSCM移入L540 (2,5 × 10^6皮下)的NSG小鼠后,观察到HL完全根除。此外,在应激L540模型中,等量的CAR30 t细胞以低、次优剂量(即50%有效t细胞剂量诱导完全缓解)输注,与输注以效应细胞为主的CAR30 t细胞相比,高富集的CAR30 TSCM (>70% TSCM)具有显著的抗肿瘤作用(引用格式:Carmen Alvarez-Fernandez, Laura Escriba-Garcia, Ana Carolina Caballero, Jorge Sierra, Javier Briones)。表达一种新型CAR30的高度富集的记忆干细胞t细胞亚群(TSCM)增强了霍奇金淋巴瘤的抗肿瘤作用[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 A023。
{"title":"Abstract A023: Highly enriched memory stem T-cell subsets (TSCM) expressing a novel CAR30 have enhanced antitumor effect in Hodgkin lymphoma","authors":"C. Álvarez-Fernández, L. Escribà-García, A. Caballero, J. Sierra, J. Briones","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A023","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A023","url":null,"abstract":"Background: Up to 20% of patients with advanced Hodgkin lymphoma (HL) are not cured with current chemo-immunotherapy treatments. Adoptive immunotherapy (AIT) with mature T-cells modified with a chimeric antigen receptor (CAR) holds promise for the treatment of various types of B cell lymphoma. The CD30 antigen, expressed in virtually all HL tumor cells, is absent in most healthy tissues, thus representing an ideal target for AIT of HL. In contrast to CAR19 for B cell malignancies, efficacy of CAR30 T-cells for Hodgkin and other CD30 lymphomas remains modest. Thus, novel approaches are needed to significantly improve the clinical efficacy of this approach. To this end, we used a novel antigen binding domain of a CD30 mAb, unaffected by soluble CD30 protein, as part of a chimeric antigen receptor (CAR) coupled to the 4.1BB and ζ chain endodomains, and transduced memory stem T-cells (TSCM) in an effort to ensure efficient engraftment, prolonged persistence, and enhancement of in vivo antitumor activity. To evaluate the feasibility of ex vivo expansion and antitumor efficacy of genetically-modified TSCM with a novel CD30-specific CAR that recognizes a membrane-proximal epitope of the CD30 molecule for the treatment of HL. Methods: A second-generation CD30-41BBz-EGFRt CAR was developed using a scFv that recognizes a membrane-proximal epitope of the CD30 protein. TSCM cells were generated with IL-7, IL-15 and IL-21, and transduced on day 2 of culture with a third-generation lentiviral vector encoding the CD30-CAR. The HL derived cell line L540 was used as tumor model. Tumor-specific cytotoxicity and cytokines were analysed at 24 hours. The in vivo antitumor efficacy was tested in a NSG (NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ) mouse model of HL xenografted with 2,5x10^6 L450 cells. Results: After 10 days, TSCM represented the majority of T-cells in culture (50.38±5.47 % for CD4+ and 70.84 ± 3.36 % for CD8+ TSCM, respectively) with a very high expression of CD30-CAR (77 ± 3.64 % in CD4+ and 83.12 ± 4.25 % in CD8+ TSCM, respectively). Expression of CD30 protein in TSCM increased over the culture period, peaking at day 8 in both CD4+ (17.5 ± 4.17 %) and CD8+ TSCM (42.37 ± 5.75 %), respectively. Despite these high levels of CD30 expression, TSCM expanded ex vivo very efficiently (CD4+ CD30-CAR: 97.24 ± 26.2 fold expansion, and CD8+ CD30-CAR: 96.79 ± 22.84 fold expansion, respectively), although lower than TSCM transduced with a CD19-CAR (CD4+ CD19-CAR: 161.96±35.9 fold expansion, and CD8+ CD19-CAR: 163.25 ± 26.51, fold expansion, respectively). Bulky CD30-CAR TSCM mediated very efficient in vitro tumor cytotoxicity (tumor cell death at 5:1 ratio: 85.25% vs. 0% with untransduced TSCM), while control CD30- targeT-cells were not killed. In addition, we evaluated the antitumor efficacy of CD30-CAR TSCM in vivo. Upon adoptive transfer of 10x10^6 bulk (CD4+ and CD8+) TSCM into NSG mice engrafted with L540 (2,5x10^6 subcutaneously), complete eradication of HL was observ","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"45 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"123918587","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 IA15: Advancing CAR T cell therapy for the treatment of brain tumors IA15: CAR - T细胞治疗脑肿瘤的进展
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-IA15
D. Alizadeh, Dongrui Wang, R. Starr, B. Aguilar, Vanessa D. Jonsson, S. Priceman, M. Barish, B. Badie, S. Forman, Christine E. Brown
Encouraging clinical experience with chimeric antigen receptor (CAR) T cells supports the notion that even immune-privileged sites such as the brain may be amenable to CAR T therapy. In the context of hematologic B cell malignancies, CD19-CARs have been shown to accumulate in the cerebrospinal fluid (CSF) and reduce the incidence of metastatic disease in the central nervous system. However, treatment of solid tumors, including brain tumors, has proven quite challenging due to heterogeneous antigen expression, suboptimal trafficking, and immunosuppressive networks in the tumor microenvironment that limit CAR T cell function and persistence. This presentation will describe our efforts to overcome these challenges for the treatment of glioblastoma (GBM), one of the most common and aggressive primary malignant brain tumors. Tumor antigen selection is a key challenge for CAR T therapy of brain tumors, as on-target/off-tumor toxicities could be life-threatening. Our preclinical studies demonstrate that a glioma-associated protein, interleukin 13 receptor alpha 2 (IL13Rα2), is an attractive CAR T cell target for glioblastoma in part because this tumor-restricted antigen is expressed by the majority of GBM. Initial clinical findings demonstrate that local delivery of IL13Rα2-CAR T cells is feasible and safe, with evidence for clinical activity in patients. In addition, as part of our on-going efforts to expand the repertoire of immunologic targets for GBM, we have optimized a HER2-CAR and developed a novel toxin-based CAR harnessing the selective GBM-binding properties of chlorotoxin (CLTX). To reduce the potential for tumor antigen escape, we are exploring approaches that leverage multiple antigens, including combining CAR T cells specific to different antigens and developing tandem CARs. Another challenge is achieving efficient tumor trafficking and infiltration of CAR T cells to brain tumors. While different routes of delivery present advantages and disadvantages, our preclinical studies using several tumor models suggest that locoregional delivery, into either the tumor or cerebral spinal fluid (CSF), is more effective than systemic delivery for treatment of brain tumors. These studies underlie our clinical approach of infusing T cells into both the resected tumor cavity and CSF. Indeed, we have demonstrated that CAR T cells administered into the CSF induced a complete response in a patient with recurrent multifocal GBM, including metastatic lesions in the spine.Additional efforts are addressing the highly immunosuppressive GBM microenvironment that limits the effectiveness of adoptively transferred T cells. Our studies highlight the impact of the PD-1/PDL-1 immune checkpoint axis in reducing the antitumor activity of CAR T cells, an inhibition that can be overcome by anti-PD-1 checkpoint blockade. In another approach designed to interrogate the interplay between the tumor microenvironment (TME), host immune system, and CAR T cells, we have establish
鼓励嵌合抗原受体(CAR) T细胞的临床经验支持这样一种观点,即即使是免疫特权部位,如大脑,也可能适合CAR T治疗。在血液B细胞恶性肿瘤的背景下,CD19-CARs已被证明在脑脊液(CSF)中积聚并降低中枢神经系统转移性疾病的发生率。然而,包括脑肿瘤在内的实体肿瘤的治疗已被证明是相当具有挑战性的,因为肿瘤微环境中的异质抗原表达、次优运输和免疫抑制网络限制了CAR - T细胞的功能和持久性。本报告将描述我们为克服胶质母细胞瘤(GBM)治疗的这些挑战所做的努力,胶质母细胞瘤是最常见和侵袭性的原发性恶性脑肿瘤之一。肿瘤抗原的选择是CAR - T治疗脑肿瘤的一个关键挑战,因为靶向/非肿瘤毒性可能危及生命。我们的临床前研究表明,一种胶质瘤相关蛋白,白细胞介素13受体α2 (IL13Rα2),是一种有吸引力的胶质母细胞瘤CAR - T细胞靶点,部分原因是这种肿瘤限制性抗原在大多数GBM中表达。初步临床结果表明,局部递送IL13Rα2-CAR T细胞是可行和安全的,并有证据表明在患者中具有临床活性。此外,作为我们持续努力扩大GBM免疫靶点的一部分,我们已经优化了HER2-CAR,并开发了一种新的基于毒素的CAR,利用氯毒素(CLTX)的选择性GBM结合特性。为了减少肿瘤抗原逃逸的可能性,我们正在探索利用多种抗原的方法,包括结合针对不同抗原的CAR - T细胞和开发串联CAR - T细胞。另一个挑战是实现肿瘤的有效运输和CAR - T细胞对脑肿瘤的浸润。虽然不同的给药途径各有利弊,但我们使用几种肿瘤模型进行的临床前研究表明,局部局部给药(进入肿瘤或脑脊液)比全身给药治疗脑肿瘤更有效。这些研究奠定了我们将T细胞注入切除肿瘤腔和脑脊液的临床方法的基础。事实上,我们已经证明CAR - T细胞进入脑脊液诱导了复发性多灶性GBM患者的完全反应,包括脊柱转移性病变。额外的努力是解决高度免疫抑制GBM微环境,限制过继转移T细胞的有效性。我们的研究强调了PD-1/PDL-1免疫检查点轴在降低CAR - T细胞抗肿瘤活性方面的影响,这种抑制可以通过抗PD-1检查点阻断来克服。在另一种旨在探究肿瘤微环境(TME)、宿主免疫系统和CAR - T细胞之间相互作用的方法中,我们建立了一个重现GBM侵袭性的同基因小鼠模型。这些研究阐明了CAR - T细胞、免疫景观和炎性TME之间的相互作用。总之,本次演讲将重点介绍目前正在进行的研究,重点是GBM的生物学、其微环境以及增强CAR - T细胞方法以更有效地治疗脑肿瘤的策略。引文格式:Darya Alizadeh, Dongrui Wang, Renate Starr, Brenda Aguilar, Vanessa D. Jonsson, Saul J. Priceman, Michael E. Barish, Behnam Badie, Stephen J. Forman, Christine E. BrownCAR - T细胞治疗脑肿瘤的进展[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1 - 15。
{"title":"Abstract IA15: Advancing CAR T cell therapy for the treatment of brain tumors","authors":"D. Alizadeh, Dongrui Wang, R. Starr, B. Aguilar, Vanessa D. Jonsson, S. Priceman, M. Barish, B. Badie, S. Forman, Christine E. Brown","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-IA15","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-IA15","url":null,"abstract":"Encouraging clinical experience with chimeric antigen receptor (CAR) T cells supports the notion that even immune-privileged sites such as the brain may be amenable to CAR T therapy. In the context of hematologic B cell malignancies, CD19-CARs have been shown to accumulate in the cerebrospinal fluid (CSF) and reduce the incidence of metastatic disease in the central nervous system. However, treatment of solid tumors, including brain tumors, has proven quite challenging due to heterogeneous antigen expression, suboptimal trafficking, and immunosuppressive networks in the tumor microenvironment that limit CAR T cell function and persistence. This presentation will describe our efforts to overcome these challenges for the treatment of glioblastoma (GBM), one of the most common and aggressive primary malignant brain tumors. Tumor antigen selection is a key challenge for CAR T therapy of brain tumors, as on-target/off-tumor toxicities could be life-threatening. Our preclinical studies demonstrate that a glioma-associated protein, interleukin 13 receptor alpha 2 (IL13Rα2), is an attractive CAR T cell target for glioblastoma in part because this tumor-restricted antigen is expressed by the majority of GBM. Initial clinical findings demonstrate that local delivery of IL13Rα2-CAR T cells is feasible and safe, with evidence for clinical activity in patients. In addition, as part of our on-going efforts to expand the repertoire of immunologic targets for GBM, we have optimized a HER2-CAR and developed a novel toxin-based CAR harnessing the selective GBM-binding properties of chlorotoxin (CLTX). To reduce the potential for tumor antigen escape, we are exploring approaches that leverage multiple antigens, including combining CAR T cells specific to different antigens and developing tandem CARs. Another challenge is achieving efficient tumor trafficking and infiltration of CAR T cells to brain tumors. While different routes of delivery present advantages and disadvantages, our preclinical studies using several tumor models suggest that locoregional delivery, into either the tumor or cerebral spinal fluid (CSF), is more effective than systemic delivery for treatment of brain tumors. These studies underlie our clinical approach of infusing T cells into both the resected tumor cavity and CSF. Indeed, we have demonstrated that CAR T cells administered into the CSF induced a complete response in a patient with recurrent multifocal GBM, including metastatic lesions in the spine.Additional efforts are addressing the highly immunosuppressive GBM microenvironment that limits the effectiveness of adoptively transferred T cells. Our studies highlight the impact of the PD-1/PDL-1 immune checkpoint axis in reducing the antitumor activity of CAR T cells, an inhibition that can be overcome by anti-PD-1 checkpoint blockade. In another approach designed to interrogate the interplay between the tumor microenvironment (TME), host immune system, and CAR T cells, we have establish","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"79 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"117252068","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 PR06: Dual-specific T-cells and an indirect vaccine eradicate large solid tumors PR06:双特异性t细胞和间接疫苗根除大实体瘤
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-PR06
C. Slaney, B. Scheidt, P. Darcy, M. Kershaw
While immunotherapy can eliminate substantial burdens of some leukemias, the ultimate challenge remains the eradication of large solid tumors and metastases for most cancers. Here we generate dual-specific T-cells expressing a chimeric antigen receptor (CAR) specific for Her2 and a TCR specific for the melanocyte protein (gp100). Injection of T-cells, together with a vaccine that contains a recombinant vaccinia virus expressing gp100, induced durable complete remission of a variety of Her2+ tumors and established metastases, some in excess of 150 mm2, in immunocompetent mice expressing Her2 in normal tissues. Tumor destruction occurred rapidly over seven days and was associated with an extensive infiltrate of T-cells. Mice that had rejected tumors were resistant to rechallenge with the same Her2+ tumor cells, indicating the formation of immune memory. Furthermore, we have established methods to transduce dual-specific T-cells from human peripheral blood with both a TCR specific for gp100 and a CAR for Her2. From as little as 1 ml of human buffy coat, we could generate sufficient numbers of cells for a course of treatment for a patient. The stimulation of gp100 through TCR enhanced the human dual-specific CAR T-cell proliferation, secretion of IFN-γ and killing of Her2+ human cancer cells in vitro. These characteristics were identified to be important for eradicating tumors in the mouse models. Taken together, our data provide valuable information for the development of CAR T-cell therapies for patients with solid cancers and evidence for pursuing a phase I clinical trial. Citation Format: Clare Y. Slaney, Bianca von Scheidt, Phillip K. Darcy, Michael H. Kershaw. Dual-specific T-cells and an indirect vaccine eradicate large solid tumors [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 PR06.
虽然免疫疗法可以消除一些白血病的大量负担,但最终的挑战仍然是根除大多数癌症的大实体瘤和转移。在这里,我们产生了双特异性t细胞,表达Her2特异性嵌合抗原受体(CAR)和黑色素细胞蛋白特异性TCR (gp100)。将t细胞与含有表达gp100的重组痘苗病毒的疫苗一起注射,在正常组织中表达Her2的免疫活性小鼠中,诱导了多种Her2+肿瘤的持久完全缓解,并建立了转移,有些转移超过150 mm2。肿瘤的破坏在7天内迅速发生,并与t细胞的广泛浸润有关。被排斥肿瘤的小鼠对同样的Her2+肿瘤细胞的再攻击有抵抗力,这表明免疫记忆的形成。此外,我们已经建立了从人外周血中转导双特异性t细胞的方法,同时具有gp100特异性TCR和Her2特异性CAR。从1毫升的人体灰褐色被毛中,我们就可以产生足够数量的细胞,用于一个病人的一个疗程。通过TCR刺激gp100增强体外人双特异性CAR - t细胞增殖、IFN-γ分泌和Her2+人癌细胞杀伤。在小鼠模型中,这些特征被认为是根除肿瘤的重要因素。综上所述,我们的数据为CAR - t细胞治疗实体癌患者的发展提供了有价值的信息,并为进行I期临床试验提供了证据。引用格式:Clare Y. Slaney, Bianca von Scheidt, Phillip K. Darcy, Michael H. Kershaw。双特异性t细胞和间接疫苗根除大实体瘤[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 PR06。
{"title":"Abstract PR06: Dual-specific T-cells and an indirect vaccine eradicate large solid tumors","authors":"C. Slaney, B. Scheidt, P. Darcy, M. Kershaw","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-PR06","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-PR06","url":null,"abstract":"While immunotherapy can eliminate substantial burdens of some leukemias, the ultimate challenge remains the eradication of large solid tumors and metastases for most cancers. Here we generate dual-specific T-cells expressing a chimeric antigen receptor (CAR) specific for Her2 and a TCR specific for the melanocyte protein (gp100). Injection of T-cells, together with a vaccine that contains a recombinant vaccinia virus expressing gp100, induced durable complete remission of a variety of Her2+ tumors and established metastases, some in excess of 150 mm2, in immunocompetent mice expressing Her2 in normal tissues. Tumor destruction occurred rapidly over seven days and was associated with an extensive infiltrate of T-cells. Mice that had rejected tumors were resistant to rechallenge with the same Her2+ tumor cells, indicating the formation of immune memory. Furthermore, we have established methods to transduce dual-specific T-cells from human peripheral blood with both a TCR specific for gp100 and a CAR for Her2. From as little as 1 ml of human buffy coat, we could generate sufficient numbers of cells for a course of treatment for a patient. The stimulation of gp100 through TCR enhanced the human dual-specific CAR T-cell proliferation, secretion of IFN-γ and killing of Her2+ human cancer cells in vitro. These characteristics were identified to be important for eradicating tumors in the mouse models. Taken together, our data provide valuable information for the development of CAR T-cell therapies for patients with solid cancers and evidence for pursuing a phase I clinical trial. Citation Format: Clare Y. Slaney, Bianca von Scheidt, Phillip K. Darcy, Michael H. Kershaw. Dual-specific T-cells and an indirect vaccine eradicate large solid tumors [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 PR06.","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"1 1-2","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"131686940","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 A032: Chimeric antigen receptor (CAR) targeted epitope determines optimal CAR spacer length for therapy against medulloblastoma 嵌合抗原受体(CAR)靶向表位决定了治疗成神经管细胞瘤的最佳CAR间隔长度
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A032
Adam J. Johnson, Cindy A. Chang, M. Baldwin, J. Yokoyama, M. Jensen
Improved therapeutic outcomes for children with embryonal brain tumors (EBTs) hinge on the development and optimization of novel, targeted therapies able to eradicate tumors without serious treatment-related damage to the central nervous system (CNS). To this end, we describe the design and validation of optimized chimeric antigen receptors (CARs) T-cells that target the EBT-associated antigen Her2. Our studies demonstrate that Her2CAR extracellular spacer domains actively influence T-cell in vitro functional outputs and in vivo responses, with the medium (M-) spacer able to confer the greatest antitumor activity against the most common EBT, medulloblastoma. Furthermore, we show that the juxtamembrane epitope targeted on Her2 precluded short spacer Her2CAR activity; yet, this activity could be rescued when the targeted epitope was expressed in a membrane distal position. Similarly, while modifications that abrogate Fc region interactions in the long spacer Her2CAR rescued in vivo activity, the resultant functional outputs failed to reach the antitumor potency elicited by the M-spacer CAR. Here we also demonstrate the in vitro and in vivo activity of M-spacer CAR T-cells produced by our clinical manufacturing process. Results from this preclinical dataset have directed the implementation of a clinical trial that delivers Her2CAR T-cells locoregionally to patients with EBT tumors, coined BrainChild-01. Collectively, these results reiterate the necessity to tailor CARs to their respective targeted antigen epitope and describe the optimization of Her2-targeted CARs for the treatment of EBT tumors. Citation Format: Adam J. Johnson, Cindy A. Chang, Michael L. Baldwin, Jason K. Yokoyama, Michael C.M. Jensen. Chimeric antigen receptor (CAR) targeted epitope determines optimal CAR spacer length for therapy against medulloblastoma [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 A032.
改善儿童胚胎性脑肿瘤(ebt)的治疗效果取决于新型靶向治疗的发展和优化,这些靶向治疗能够根除肿瘤而不会对中枢神经系统(CNS)造成严重的治疗相关损伤。为此,我们描述了优化嵌合抗原受体(CARs) t细胞的设计和验证,靶向ebt相关抗原Her2。我们的研究表明,Her2CAR细胞外间隔结构域积极影响t细胞的体外功能输出和体内反应,中等(M-)间隔结构域能够对最常见的EBT、成神经管细胞瘤赋予最大的抗肿瘤活性。此外,我们发现靶向Her2的近膜表位阻止了短间隔Her2CAR的活性;然而,当目标表位在膜的远端位置表达时,这种活性可以被挽救。类似地,虽然在长间隔段Her2CAR中去除Fc区相互作用的修饰挽救了体内活性,但由此产生的功能输出未能达到m间隔段CAR所激发的抗肿瘤效力。在这里,我们还展示了通过我们的临床制造过程产生的m间隔CAR - t细胞的体外和体内活性。该临床前数据集的结果指导了一项临床试验的实施,该试验将Her2CAR - t细胞局部递送给EBT肿瘤患者,称为BrainChild-01。总的来说,这些结果重申了根据各自的靶向抗原表位定制car的必要性,并描述了her2靶向car治疗EBT肿瘤的优化。引用格式:Adam J. Johnson, Cindy A. Chang, Michael L. Baldwin, Jason K. Yokoyama, Michael C.M. Jensen。嵌合抗原受体(CAR)靶向表位决定了治疗成神经管细胞瘤的最佳CAR间隔长度[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 A032。
{"title":"Abstract A032: Chimeric antigen receptor (CAR) targeted epitope determines optimal CAR spacer length for therapy against medulloblastoma","authors":"Adam J. Johnson, Cindy A. Chang, M. Baldwin, J. Yokoyama, M. Jensen","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A032","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A032","url":null,"abstract":"Improved therapeutic outcomes for children with embryonal brain tumors (EBTs) hinge on the development and optimization of novel, targeted therapies able to eradicate tumors without serious treatment-related damage to the central nervous system (CNS). To this end, we describe the design and validation of optimized chimeric antigen receptors (CARs) T-cells that target the EBT-associated antigen Her2. Our studies demonstrate that Her2CAR extracellular spacer domains actively influence T-cell in vitro functional outputs and in vivo responses, with the medium (M-) spacer able to confer the greatest antitumor activity against the most common EBT, medulloblastoma. Furthermore, we show that the juxtamembrane epitope targeted on Her2 precluded short spacer Her2CAR activity; yet, this activity could be rescued when the targeted epitope was expressed in a membrane distal position. Similarly, while modifications that abrogate Fc region interactions in the long spacer Her2CAR rescued in vivo activity, the resultant functional outputs failed to reach the antitumor potency elicited by the M-spacer CAR. Here we also demonstrate the in vitro and in vivo activity of M-spacer CAR T-cells produced by our clinical manufacturing process. Results from this preclinical dataset have directed the implementation of a clinical trial that delivers Her2CAR T-cells locoregionally to patients with EBT tumors, coined BrainChild-01. Collectively, these results reiterate the necessity to tailor CARs to their respective targeted antigen epitope and describe the optimization of Her2-targeted CARs for the treatment of EBT tumors. Citation Format: Adam J. Johnson, Cindy A. Chang, Michael L. Baldwin, Jason K. Yokoyama, Michael C.M. Jensen. Chimeric antigen receptor (CAR) targeted epitope determines optimal CAR spacer length for therapy against medulloblastoma [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 A032.","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"116 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"133279237","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 A041: Hypoxia-responsive CAR T-cells 缺氧反应性CAR - t细胞
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A041
Tina Sarén, Anne Marie Senz, Mohanraj Ramachandran, Di Yu, M. Essand
In recent years, CD19 CAR T-cell therapy has been successfully implemented against therapy-resistant B-cell malignancies. The results are especially striking for acute lymphoblastic leukemia (ALL), while results are somewhat less stunning when it comes to lymphoma. This is in part due to the semi-solid structure of lymphoma with an immune suppressive tumor microenvironment and less oxygenated areas within the tumors. These obstacles become even more pronounced in solid tumors. Furthermore, in the case of solid tumors the lack of tumor-specific antigens represents a major challenge. Instead, overexpressed tumor-associated antigens (TAAs) are often used as targets in CAR T-cell therapy of solid tumors. However, as TAAs are also expressed to some extent in normal tissue, targeting these antigens may result in severe OFF-tumor ON-target toxicity. In the case of CD19 CAR T-cell therapy of B-cell malignancies, OFF-tumor ON-target toxicity causes B-cell aplasia.The aim of this study was to reduce OFF-tumor ON-target toxicity by engineering CAR T-cells that mainly express the CAR molecule in the hypoxic tumor microenvironment. Hypoxia-inducible factor 1 (HIF1) is a transcription factor that is a major mediator of hypoxia-induced gene expression as it binds to hypoxia response elements (HREs) in promoter regions of hypoxia-responsive genes and initiates their expression. In this study, a cassette of HREs was inserted together with a minimal CMV (mCMV) promoter in front of the CD19 CAR cassette to promote expression during hypoxic conditions. Chemically induced hypoxia augmented CD19 CAR expression over time in hypoxia-responsive CD19 CAR T-cells (HRE-mCMV-CD19CAR-GFP), whereas CAR expression was unaffected in CD19 CAR T-cells with constitutively expressed CAR (EF1α-CD19CAR-GFP). Furthermore, hypoxia-responsive CAR T-cells had an increased activation status and killing capacity upon antigen encounter in hypoxic compared to normoxic conditions. This design could be evaluated in the clinic to reduce B-cell aplasia in CD19 CAR T-cell therapy of lymphomas and especially to reduce OFF-target toxicities from CAR T-cells targeting TAAs in solid tumors. Citation Format: Tina Anna Saren, Anne Marie Senz, Mohanraj Ramachandran, Di Yu, Magnus Essand. Hypoxia-responsive CAR T-cells [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 A041.
近年来,CD19 CAR - t细胞疗法已经成功地用于治疗耐药的b细胞恶性肿瘤。对于急性淋巴细胞白血病(ALL)的结果尤其惊人,而对于淋巴瘤的结果则不那么惊人。这部分是由于淋巴瘤的半实体结构具有免疫抑制的肿瘤微环境和肿瘤内较少的含氧区域。这些障碍在实体瘤中变得更加明显。此外,在实体瘤的情况下,缺乏肿瘤特异性抗原是一个主要的挑战。相反,过度表达的肿瘤相关抗原(TAAs)通常被用作实体瘤CAR - t细胞治疗的靶点。然而,由于TAAs在正常组织中也有一定程度的表达,靶向这些抗原可能会导致严重的OFF-tumor ON-target毒性。在CD19 CAR -t细胞治疗b细胞恶性肿瘤的情况下,OFF-tumor ON-target毒性导致b细胞发育不全。本研究的目的是通过在低氧肿瘤微环境中设计主要表达CAR分子的CAR -t细胞来降低off -肿瘤ON-target毒性。缺氧诱导因子1 (hypoxia- inducible factor 1, HIF1)是一种转录因子,是缺氧诱导基因表达的主要媒介,因为它与缺氧反应基因启动子区域的缺氧反应元件(hypoxia response elements, HREs)结合并启动其表达。在这项研究中,将HREs盒与最小CMV (mCMV)启动子一起插入CD19 CAR盒前,以促进缺氧条件下的表达。随着时间的推移,化学诱导的缺氧在低氧应答的CD19CAR- t细胞(HRE-mCMV-CD19CAR-GFP)中增强了CD19CAR的表达,而在组成型表达CAR的CD19CAR- t细胞(EF1α-CD19CAR-GFP)中,CAR的表达不受影响。此外,与常氧条件相比,低氧反应的CAR - t细胞在低氧条件下遇到抗原时具有更高的激活状态和杀伤能力。这种设计可以在临床上进行评估,以减少CD19 CAR -t细胞治疗淋巴瘤中的b细胞发育不全,特别是减少实体瘤中靶向TAAs的CAR -t细胞的脱靶毒性。引文格式:Tina Anna Saren, Anne Marie Senz, Mohanraj Ramachandran, Di Yu, Magnus Essand。低氧反应CAR - t细胞[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 A041。
{"title":"Abstract A041: Hypoxia-responsive CAR T-cells","authors":"Tina Sarén, Anne Marie Senz, Mohanraj Ramachandran, Di Yu, M. Essand","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A041","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A041","url":null,"abstract":"In recent years, CD19 CAR T-cell therapy has been successfully implemented against therapy-resistant B-cell malignancies. The results are especially striking for acute lymphoblastic leukemia (ALL), while results are somewhat less stunning when it comes to lymphoma. This is in part due to the semi-solid structure of lymphoma with an immune suppressive tumor microenvironment and less oxygenated areas within the tumors. These obstacles become even more pronounced in solid tumors. Furthermore, in the case of solid tumors the lack of tumor-specific antigens represents a major challenge. Instead, overexpressed tumor-associated antigens (TAAs) are often used as targets in CAR T-cell therapy of solid tumors. However, as TAAs are also expressed to some extent in normal tissue, targeting these antigens may result in severe OFF-tumor ON-target toxicity. In the case of CD19 CAR T-cell therapy of B-cell malignancies, OFF-tumor ON-target toxicity causes B-cell aplasia.The aim of this study was to reduce OFF-tumor ON-target toxicity by engineering CAR T-cells that mainly express the CAR molecule in the hypoxic tumor microenvironment. Hypoxia-inducible factor 1 (HIF1) is a transcription factor that is a major mediator of hypoxia-induced gene expression as it binds to hypoxia response elements (HREs) in promoter regions of hypoxia-responsive genes and initiates their expression. In this study, a cassette of HREs was inserted together with a minimal CMV (mCMV) promoter in front of the CD19 CAR cassette to promote expression during hypoxic conditions. Chemically induced hypoxia augmented CD19 CAR expression over time in hypoxia-responsive CD19 CAR T-cells (HRE-mCMV-CD19CAR-GFP), whereas CAR expression was unaffected in CD19 CAR T-cells with constitutively expressed CAR (EF1α-CD19CAR-GFP). Furthermore, hypoxia-responsive CAR T-cells had an increased activation status and killing capacity upon antigen encounter in hypoxic compared to normoxic conditions. This design could be evaluated in the clinic to reduce B-cell aplasia in CD19 CAR T-cell therapy of lymphomas and especially to reduce OFF-target toxicities from CAR T-cells targeting TAAs in solid tumors. Citation Format: Tina Anna Saren, Anne Marie Senz, Mohanraj Ramachandran, Di Yu, Magnus Essand. Hypoxia-responsive CAR T-cells [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 A041.","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"70 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"121786037","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 A037: A novel pharmacologic “ON/OFF” switch to modulate CAR-T-cell function in vitro and in vivo 摘要:一种新的药理“ON/OFF”开关在体内和体外调节car - t细胞功能
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A037
K. Mestermann, R. Julian, Frenz Silke, E. Hermann, M. Hudecek
Background: Immunotherapy with CAR-T-cells (CAR-T) is a powerful novel treatment for hematologic malignancies, but also bound with significant acute and chronic side effects, including potentially life-threatening cytokine release syndrome (CRS) and on-target recognition of normal cells expressing the targeted antigen. This toxicity limits clinical utility and is at least in part caused by the inability to effectively control CAR-T function following infusion. Here, we present a novel strategy of pharmacologic “ON/OFF” switch to precisely control CAR-T function in real-time, which we demonstrate to modulate T-cell activity in vitro and in vivo. Methods: We considered that an effective way for controlling CAR-T function was to interfere with signal transduction though the CAR. We assembled a library of clinically approved drug compounds and screened for their ability to reversible block CAR-T function without affecting CAR-T viability. We performed functional testing with CD8+ and CD4+ CAR-T (n=3 donors) in the presence of titrated doses of the lead compound, and employed CD19- and ROR1-specific CARs comprising 4-1BB or CD28 costimulatory moieties. Results: We identified a lead compound, TCI-1, that stood out through its ability to confer a dose-dependent (partial at lower, complete at higher doses) blockade of all CAR-T effector functions, i.e., cytolytic activity, cytokine secretion and proliferation. We confirmed TCI-1 was effective in both CD8+ and CD4+ T-cells, and in each of the 3 CAR constructs. The onset of CAR-T blockade was immediate after exposure to TCI-1 and was caused by interference with early phosphorylation events in the CAR signaling cascade as demonstrated by Western blot, and interference with the induction of transcription factors, as demonstrated with an NFAT-inducible reporter gene. Intriguingly, blockade of CAR-T function was effective for several days if exposure to TCI-1 was sustained and instantaneously and fully reversible after removal of the compound. Short- and long-term exposure to TCI-1 did not induce a reduction of CAR-T viability, and did not hinder the subsequent ability of CAR-T to exert their functions. We considered that in patients with CRS, CAR-T are in an activated state, and performed comprehensive testing to show that TCI-1 was able to arrest CAR-T that are in the process of executing their effector functions. In addition, we employed a xenograft model in immunodeficient mice (NSG/Raji) to determine whether TCI-1 was capable of controlling the function of CD19 CAR-T-cells in vivo. Indeed, we demonstrate that administration of TCI-1 conferred a functional arrest of CAR-T, and that CAR-T resumed their antitumor function once administration of TCI-1 was discontinued. Conclusions: Our data show that TCI-1 is capable to exert real-time, on/off control over CAR-T function, suggesting the potential to prevent or mitigate side effects of CAR-T therapy in a clinical setting. The reversible complete inhibition of
背景:CAR-T细胞免疫疗法(CAR-T)是治疗血液系统恶性肿瘤的一种强有力的新疗法,但也伴随着严重的急性和慢性副作用,包括潜在的危及生命的细胞因子释放综合征(CRS)和表达靶向抗原的正常细胞的靶向识别。这种毒性限制了临床应用,至少部分是由于输注后无法有效控制CAR-T功能造成的。在这里,我们提出了一种新的药物“ON/OFF”开关策略来实时精确控制CAR-T功能,我们证明了它在体外和体内都能调节t细胞的活性。方法:我们认为通过CAR干扰信号转导是控制CAR- t功能的有效途径。我们组装了一个临床批准的药物化合物库,并筛选了它们在不影响CAR-T生存能力的情况下可逆阻断CAR-T功能的能力。我们用CD8+和CD4+ CAR-T (n=3个供体)在先导化合物的滴定剂量下进行了功能测试,并使用了含有4-1BB或CD28共刺激部分的CD19和ror1特异性CAR-T。结果:我们确定了一种先导化合物TCI-1,它具有剂量依赖性(低剂量时部分阻断,高剂量时完全阻断),能够阻断所有CAR-T效应功能,即细胞溶解活性、细胞因子分泌和增殖。我们证实TCI-1在CD8+和CD4+ t细胞以及3种CAR结构中的每一种中都有效。暴露于TCI-1后立即发生CAR- t阻断,其原因是经Western blot证实,CAR- t信号级联中的早期磷酸化事件受到干扰,转录因子的诱导受到干扰,经nfat诱导的报告基因证实。有趣的是,如果持续暴露于TCI-1,并且在去除化合物后立即完全可逆,那么CAR-T功能的阻断在几天内是有效的。短期和长期暴露于TCI-1不会导致CAR-T活性降低,也不会阻碍CAR-T随后发挥其功能的能力。我们认为在CRS患者中,CAR-T处于激活状态,并进行了全面的测试,以证明TCI-1能够阻止正在执行其效应功能的CAR-T。此外,我们采用免疫缺陷小鼠(NSG/Raji)异种移植模型来确定TCI-1是否能够在体内控制CD19 car - t细胞的功能。事实上,我们证明了TCI-1的施用可以使CAR-T功能阻滞,并且一旦TCI-1的施用停止,CAR-T恢复其抗肿瘤功能。结论:我们的数据显示,TCI-1能够对CAR-T功能施加实时的开/关控制,这表明在临床环境中有可能预防或减轻CAR-T治疗的副作用。通过TCI-1对CAR-T功能的可逆完全抑制,而不损害CAR-T的生存能力,超过了对t细胞有毒且仅提供不完全功能控制的类固醇的质量,并补充了自杀基因策略,有效控制慢性副作用,但也取消了CAR-T的抗肿瘤作用。引文格式:Katrin Mestermann, Rydzek Julian, Frenz Silke, Einsele Hermann, Michael Hudecek。一种新型的药理学“ON/OFF”开关在体外和体内调节car - t细胞功能[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 A037。
{"title":"Abstract A037: A novel pharmacologic “ON/OFF” switch to modulate CAR-T-cell function in vitro and in vivo","authors":"K. Mestermann, R. Julian, Frenz Silke, E. Hermann, M. Hudecek","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A037","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A037","url":null,"abstract":"Background: Immunotherapy with CAR-T-cells (CAR-T) is a powerful novel treatment for hematologic malignancies, but also bound with significant acute and chronic side effects, including potentially life-threatening cytokine release syndrome (CRS) and on-target recognition of normal cells expressing the targeted antigen. This toxicity limits clinical utility and is at least in part caused by the inability to effectively control CAR-T function following infusion. Here, we present a novel strategy of pharmacologic “ON/OFF” switch to precisely control CAR-T function in real-time, which we demonstrate to modulate T-cell activity in vitro and in vivo. Methods: We considered that an effective way for controlling CAR-T function was to interfere with signal transduction though the CAR. We assembled a library of clinically approved drug compounds and screened for their ability to reversible block CAR-T function without affecting CAR-T viability. We performed functional testing with CD8+ and CD4+ CAR-T (n=3 donors) in the presence of titrated doses of the lead compound, and employed CD19- and ROR1-specific CARs comprising 4-1BB or CD28 costimulatory moieties. Results: We identified a lead compound, TCI-1, that stood out through its ability to confer a dose-dependent (partial at lower, complete at higher doses) blockade of all CAR-T effector functions, i.e., cytolytic activity, cytokine secretion and proliferation. We confirmed TCI-1 was effective in both CD8+ and CD4+ T-cells, and in each of the 3 CAR constructs. The onset of CAR-T blockade was immediate after exposure to TCI-1 and was caused by interference with early phosphorylation events in the CAR signaling cascade as demonstrated by Western blot, and interference with the induction of transcription factors, as demonstrated with an NFAT-inducible reporter gene. Intriguingly, blockade of CAR-T function was effective for several days if exposure to TCI-1 was sustained and instantaneously and fully reversible after removal of the compound. Short- and long-term exposure to TCI-1 did not induce a reduction of CAR-T viability, and did not hinder the subsequent ability of CAR-T to exert their functions. We considered that in patients with CRS, CAR-T are in an activated state, and performed comprehensive testing to show that TCI-1 was able to arrest CAR-T that are in the process of executing their effector functions. In addition, we employed a xenograft model in immunodeficient mice (NSG/Raji) to determine whether TCI-1 was capable of controlling the function of CD19 CAR-T-cells in vivo. Indeed, we demonstrate that administration of TCI-1 conferred a functional arrest of CAR-T, and that CAR-T resumed their antitumor function once administration of TCI-1 was discontinued. Conclusions: Our data show that TCI-1 is capable to exert real-time, on/off control over CAR-T function, suggesting the potential to prevent or mitigate side effects of CAR-T therapy in a clinical setting. The reversible complete inhibition of ","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"59 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"115943101","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 A039: FAM49B–specific regulatory T-cells recognize and target cancer cells in the context of Qa-1 A039: fam49b特异性调节性t细胞在Qa-1的背景下识别和靶向癌细胞
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A039
H. Nakagawa, Hye-Jung Kim, H. Cantor
The outcome of cancer patients has recently been improved by utilizing novel immunotherapies that inhibit the regulatory immune system. Regulatory T-cells (Treg) contribute not only to suppression of excessive immune responses in the steady state but also to reduction of antitumor immune responses in cancer patients. We have shown that down-regulation of the Helios transcription factor undermines CD4 Treg stability inducing these cells to convert into effector cells that enhance the antitumor immune response. Helios is also the canonical transcription factor of CD8 Treg and essential for CD8 Treg function. CD8 Treg are Qa-1 (HLA-E in human)–restricted CD8 T-cells that mainly regulate Qa-1+ immune cells. The peptide FAM49B(p190-198) is a Qa-1 binding peptide derived from ubiquitously expressed FAM49B. Although FAM49B(p190-198) is regularly degenerated in the endoplasmic reticulum normal cells, we found that FAM49B(p190-198) is stably presented in some tumor cells. Here, we investigate the potential contribution of FAM49B(p190-198)/Qa-1 specific CD8 Treg to cancer.FAM49B(p190-198)/Qa-1 specific CD8 Treg were detected by FAM49B(p190-198)/Qa-1 tetramer after immunization with FAM49B(p190-198) peptide–loaded dendritic cells in CD8 Treg (i.e., CD44+CD122+Ly49+ CD8 T-cells). T-cell receptor cDNAs were amplified from the sorted single cells and inserted into retrovirus expression vectors for transduction in 58α–β– hybridoma cells and bone marrow cells. EL4-Qa-1 KO cells and EL4-FAM49B KO cells were generated using CRISPR/Cas9 technology. These tools allowed validation of the specificity of the TCRs and their Qa-1–restriction. For in vivo studies, TCR retrogenic mice were developed by transferring TCR-transduced Rag2–/– bone marrow hematopoietic stem cells into lethally irradiated CD45.1 mice. Retrogenic T-cells were prepared by CD8 enrichment of retrogenic mouse splenocytes and in vitro culture for 3 days in the presence of IL15c. For in vivo studies, we use an adoptive transfer model of retrogenic T-cells for EL4 tumor bearing mice. Rag2–/–γC–/– mice were inoculated with 0.5 x 10^6 EL4 WT-cells and 0.5 x 10^6 EL4 Qa-1 KO cells on each lateral flank, then intravenously injected with retrogenic T-cells (on day 13 and 17). FAM49B(p190-198)/Qa-1 specific CD8 T-cells preferentially use TCR Vα3.2 (TRAV9N-3) and Vβ5.1/5.2 (TRBV12-1/12-2) chains independent of their expression of Ly49+. We tested 17 pairs obtained from Ly49+ cells using the in vitro system and found that 11 pairs bound strongly to Fam49b(p190-198)/Qa-1 tetramer while 8 out of the 11 pairs exhibited antigen-specific activation. Interestingly, one TCR pair (No.8) responded to WT EL4 cells without antigen peptides, which was abolished in coculture wells with Qa-1 KO EL4 cells and FAM49B KO EL4 cells. This TCR hybridoma was also activated by stimulated WT B6 splenocytes but not by Qa-1 KO splenocytes suggesting its regulatory potential. Adoptive transfer of TCR No.8-expressing retrogenic T-cells de
癌症患者的治疗结果最近通过使用抑制调节免疫系统的新型免疫疗法得到了改善。调节性t细胞(Regulatory t cells, Treg)不仅在稳定状态下抑制过度的免疫反应,而且还有助于降低癌症患者的抗肿瘤免疫反应。我们已经证明Helios转录因子的下调会破坏CD4 Treg的稳定性,诱导这些细胞转化为增强抗肿瘤免疫反应的效应细胞。Helios也是CD8 Treg的典型转录因子,对CD8 Treg的功能至关重要。CD8 Treg是Qa-1(人HLA-E)限制性CD8 t细胞,主要调控Qa-1+免疫细胞。肽FAM49B(p190-198)是由普遍表达的FAM49B衍生的Qa-1结合肽。虽然FAM49B(p190-198)在内质网正常细胞中有规律地变性,但我们发现FAM49B(p190-198)在一些肿瘤细胞中稳定存在。在此,我们研究了FAM49B(p190-198)/Qa-1特异性CD8 Treg对癌症的潜在贡献。用FAM49B(p190-198)/Qa-1四聚体免疫CD8 Treg(即CD44+CD122+Ly49+ CD8 t细胞)后,FAM49B(p190-198)/Qa-1特异性CD8 Treg检测FAM49B(p190-198)/Qa-1四聚体。从分选的单细胞中扩增t细胞受体cdna,插入逆转录病毒表达载体,在58α - β -杂交瘤细胞和骨髓细胞中转导。采用CRISPR/Cas9技术制备EL4-Qa-1 KO细胞和EL4-FAM49B KO细胞。这些工具允许验证tcr的特异性及其qa -1限制。在体内研究中,通过将TCR转导的Rag2 - / -骨髓造血干细胞转移到致死辐照的CD45.1小鼠中,培养TCR逆转录小鼠。用CD8富集逆转录小鼠脾细胞,在il - 15c存在下体外培养3天,制备逆转录t细胞。在体内研究中,我们使用了EL4荷瘤小鼠的逆转录t细胞过继转移模型。Rag2 - / - γ c - / -小鼠分别接种0.5 × 10^6 EL4 wt细胞和0.5 × 10^6 EL4 Qa-1 KO细胞,然后静脉注射逆转录t细胞(第13天和第17天)。FAM49B(p190-198)/Qa-1特异性CD8 t细胞优先使用独立于Ly49+表达的TCR Vα3.2 (TRAV9N-3)和Vβ5.1/5.2 (TRBV12-1/12-2)链。我们使用体外系统测试了从Ly49+细胞中获得的17对,发现11对与Fam49b(p190-198)/Qa-1四聚体结合强烈,11对中有8对表现出抗原特异性活化。有趣的是,一个TCR对(No.8)对没有抗原肽的WT EL4细胞有反应,在与Qa-1 KO EL4细胞和FAM49B KO EL4细胞共培养孔中被消除。这种TCR杂交瘤也能被WT B6脾细胞激活,但不能被Qa-1 KO脾细胞激活,这表明它具有调节潜力。表达TCR no .8的逆转录t细胞的过继转移在同一宿主中接种时,延迟了EL4 WT肿瘤的生长,但没有延迟EL4 Qa-1 KO肿瘤的生长。与KO肿瘤相比,肿瘤浸润性CD8 t细胞在WT肿瘤中表现出更活化的表型,表明逆转录t细胞以qa -1依赖的方式识别和靶向EL4细胞。引文格式:Hidetoshi Nakagawa, Hye-Jung Kim, Harvey Cantor。fam49b特异性调节性t细胞在Qa-1的背景下识别和靶向癌细胞[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 A039。
{"title":"Abstract A039: FAM49B–specific regulatory T-cells recognize and target cancer cells in the context of Qa-1","authors":"H. Nakagawa, Hye-Jung Kim, H. Cantor","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A039","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A039","url":null,"abstract":"The outcome of cancer patients has recently been improved by utilizing novel immunotherapies that inhibit the regulatory immune system. Regulatory T-cells (Treg) contribute not only to suppression of excessive immune responses in the steady state but also to reduction of antitumor immune responses in cancer patients. We have shown that down-regulation of the Helios transcription factor undermines CD4 Treg stability inducing these cells to convert into effector cells that enhance the antitumor immune response. Helios is also the canonical transcription factor of CD8 Treg and essential for CD8 Treg function. CD8 Treg are Qa-1 (HLA-E in human)–restricted CD8 T-cells that mainly regulate Qa-1+ immune cells. The peptide FAM49B(p190-198) is a Qa-1 binding peptide derived from ubiquitously expressed FAM49B. Although FAM49B(p190-198) is regularly degenerated in the endoplasmic reticulum normal cells, we found that FAM49B(p190-198) is stably presented in some tumor cells. Here, we investigate the potential contribution of FAM49B(p190-198)/Qa-1 specific CD8 Treg to cancer.FAM49B(p190-198)/Qa-1 specific CD8 Treg were detected by FAM49B(p190-198)/Qa-1 tetramer after immunization with FAM49B(p190-198) peptide–loaded dendritic cells in CD8 Treg (i.e., CD44+CD122+Ly49+ CD8 T-cells). T-cell receptor cDNAs were amplified from the sorted single cells and inserted into retrovirus expression vectors for transduction in 58α–β– hybridoma cells and bone marrow cells. EL4-Qa-1 KO cells and EL4-FAM49B KO cells were generated using CRISPR/Cas9 technology. These tools allowed validation of the specificity of the TCRs and their Qa-1–restriction. For in vivo studies, TCR retrogenic mice were developed by transferring TCR-transduced Rag2–/– bone marrow hematopoietic stem cells into lethally irradiated CD45.1 mice. Retrogenic T-cells were prepared by CD8 enrichment of retrogenic mouse splenocytes and in vitro culture for 3 days in the presence of IL15c. For in vivo studies, we use an adoptive transfer model of retrogenic T-cells for EL4 tumor bearing mice. Rag2–/–γC–/– mice were inoculated with 0.5 x 10^6 EL4 WT-cells and 0.5 x 10^6 EL4 Qa-1 KO cells on each lateral flank, then intravenously injected with retrogenic T-cells (on day 13 and 17). FAM49B(p190-198)/Qa-1 specific CD8 T-cells preferentially use TCR Vα3.2 (TRAV9N-3) and Vβ5.1/5.2 (TRBV12-1/12-2) chains independent of their expression of Ly49+. We tested 17 pairs obtained from Ly49+ cells using the in vitro system and found that 11 pairs bound strongly to Fam49b(p190-198)/Qa-1 tetramer while 8 out of the 11 pairs exhibited antigen-specific activation. Interestingly, one TCR pair (No.8) responded to WT EL4 cells without antigen peptides, which was abolished in coculture wells with Qa-1 KO EL4 cells and FAM49B KO EL4 cells. This TCR hybridoma was also activated by stimulated WT B6 splenocytes but not by Qa-1 KO splenocytes suggesting its regulatory potential. Adoptive transfer of TCR No.8-expressing retrogenic T-cells de","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"43 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"115658748","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
Genetically Engineered 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