Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-IA14
S. Rosenberg
Adoptive cell transfer (ACT) immunotherapy using T lymphocytes with antitumor activity is a living therapy that can be highly effective in patients with a variety of metastatic cancers. Naturally occurring tumor infiltrating lymphocytes (TIL) expanded in vitro and administered following a lymphodepleting preparative regimen mediated objective regression of metastatic melanoma in 55% of 194 patients including complete regressions in 24% of patients who remain ongoing disease-free 3 to 10 years later. To determine the antigens recognized by TIL we developed an approach based on deep exomic sequencing of the cancer and immunologic testing of TIL or peripheral lymphocytes to generate T-cells that recognized immunogenic mutations. TIL from 22 patients with metastatic melanoma recognized 54 random somatic mutations none of which were shared among different melanomas. We next extended these studies to patients with common epithelial cancers and showed that 81 of 99 (82%) patients with a variety human cancer types including esophageal, colorectal, bile duct, gastric, pancreatic, ovarian, cervical and lung cancer contained T-cells that recognized 197 neoantigens all of which were unique except for KRAS (2 patients). Targeting unique cancer mutations has extended the reach of ACT immunotherapy and was used to mediate objective regressions in selected patients with chemorefractory metastatic cancers of the bile duct, colon, cervix, and breast. In addition to the use of naturally occurring mutation-reactive cells, we have genetically engineered autologous lymphocytes to express shared antitumor T- cell receptors (TCR) or chimeric antigen receptors (CAR) for use in ACT immunotherapy that can mediate durable cancer regressions in heavily pretreated patients with refractory lymphomas, sarcomas and melanoma. Autologous T- cells can be used to provide a highly personalized immunotherapy for cancer patients refractory to conventional cancer treatments. Citation Format: Steven A. Rosenberg. Cell transfer immunotherapy targeting unique somatic mutations in cancer [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr IA14.
使用具有抗肿瘤活性的T淋巴细胞的过继细胞转移(ACT)免疫疗法是一种对各种转移性癌症患者非常有效的活疗法。自然发生的肿瘤浸润淋巴细胞(TIL)在体外扩大,并在淋巴细胞消耗预备方案后给予治疗,介导了194例患者中55%的转移性黑色素瘤的客观消退,其中24%的患者在3至10年后保持无病状态,完全消退。为了确定TIL识别的抗原,我们开发了一种基于癌症的深度外显子组测序和TIL或外周淋巴细胞的免疫学测试来产生识别免疫原性突变的t细胞的方法。来自22名转移性黑色素瘤患者的TIL识别出54个随机体细胞突变,其中没有一个在不同的黑色素瘤中共享。接下来,我们将这些研究扩展到常见上皮癌患者,结果显示,99例(82%)多种人类癌症类型(包括食管癌、结直肠癌、胆管癌、胃癌、胰腺癌、卵巢癌、宫颈癌和肺癌)患者中有81例(82%)含有识别197种新抗原的t细胞,除了KRAS(2例)外,所有这些新抗原都是独一无二的。针对独特的癌症突变已经扩展了ACT免疫治疗的范围,并被用于介导胆管、结肠癌、宫颈癌和乳腺癌化疗难治性转移癌患者的客观回归。除了使用自然发生的突变反应细胞外,我们还利用基因工程的自体淋巴细胞来表达共享的抗肿瘤T细胞受体(TCR)或嵌合抗原受体(CAR),用于ACT免疫治疗,可以介导重度预处理的难治性淋巴瘤、肉瘤和黑色素瘤患者的持久癌症消退。自体T细胞可以为难以接受常规癌症治疗的癌症患者提供高度个性化的免疫治疗。引文格式:Steven A. Rosenberg。针对癌症中独特体细胞突变的细胞转移免疫治疗[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11 - 14。
{"title":"Abstract IA14: Cell transfer immunotherapy targeting unique somatic mutations in cancer","authors":"S. Rosenberg","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-IA14","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-IA14","url":null,"abstract":"Adoptive cell transfer (ACT) immunotherapy using T lymphocytes with antitumor activity is a living therapy that can be highly effective in patients with a variety of metastatic cancers. Naturally occurring tumor infiltrating lymphocytes (TIL) expanded in vitro and administered following a lymphodepleting preparative regimen mediated objective regression of metastatic melanoma in 55% of 194 patients including complete regressions in 24% of patients who remain ongoing disease-free 3 to 10 years later. To determine the antigens recognized by TIL we developed an approach based on deep exomic sequencing of the cancer and immunologic testing of TIL or peripheral lymphocytes to generate T-cells that recognized immunogenic mutations. TIL from 22 patients with metastatic melanoma recognized 54 random somatic mutations none of which were shared among different melanomas. We next extended these studies to patients with common epithelial cancers and showed that 81 of 99 (82%) patients with a variety human cancer types including esophageal, colorectal, bile duct, gastric, pancreatic, ovarian, cervical and lung cancer contained T-cells that recognized 197 neoantigens all of which were unique except for KRAS (2 patients). Targeting unique cancer mutations has extended the reach of ACT immunotherapy and was used to mediate objective regressions in selected patients with chemorefractory metastatic cancers of the bile duct, colon, cervix, and breast. In addition to the use of naturally occurring mutation-reactive cells, we have genetically engineered autologous lymphocytes to express shared antitumor T- cell receptors (TCR) or chimeric antigen receptors (CAR) for use in ACT immunotherapy that can mediate durable cancer regressions in heavily pretreated patients with refractory lymphomas, sarcomas and melanoma. Autologous T- cells can be used to provide a highly personalized immunotherapy for cancer patients refractory to conventional cancer treatments. Citation Format: Steven A. Rosenberg. Cell transfer immunotherapy targeting unique somatic mutations in cancer [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr IA14.","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"2 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":"125149609","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-PR05
C. Klebanoff
Across clinical trials, T-cell expansion and persistence following adoptive cell transfer (ACT) has correlated with superior patient outcomes. Herein, we undertook a pan-cancer analysis across human tumors to identify potentially actionable ligand/receptor pairs that might limit T-cell function and persistence following ACT. We discovered that FASLG, the gene encoding the apoptosis-inducing ligand FasL, is overexpressed within the majority of human tumor microenvironments. Further, we uncovered that Fas, the receptor for FasL, is highly expressed on patient-derived T-cells used for ACT. We hypothesized that a cognate Fas-FasL interaction within the tumor microenvironment might limit both T-cell persistence and antitumor efficacy. We discovered that genetic engineering of Fas variants impaired the ability to bind FADD function as dominant negative receptors (DNRs) in Fas-competent mouse and human T-cells, rescuing cells from FasL-induced apoptosis. Fas DNR-engineered T-cells exhibited enhanced persistence within tumors following ACT, resulting in superior cancer regression and overall survival in solid and hematologic malignancies treated with TCR or CAR-modified cells. Despite enhanced longevity, Fas DNR-engineered T-cells did not undergo aberrant clonal expansion, demonstrating the safety of this approach. Thus, cell-intrinsic “insulation” of T-cells from the negative influence of FasL is a potentially universal strategy to enhance ACT efficacy across a broad range of human malignancies. Citation Format: Christopher A. Klebanoff. T-cells engineered to overcome death signaling within the tumor microenvironment enhance adoptive cancer immunotherapy [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr PR05.
在临床试验中,过继细胞移植(ACT)后的t细胞扩增和持续存在与患者的良好预后相关。在此,我们对人类肿瘤进行了泛癌症分析,以确定可能限制ACT后t细胞功能和持久性的潜在可操作的配体/受体对。我们发现,FASLG,编码凋亡诱导配体FasL的基因,在大多数人类肿瘤微环境中过表达。此外,我们发现FasL的受体Fas在用于ACT的患者源性t细胞上高度表达。我们假设肿瘤微环境中同源的fasl - fasl相互作用可能限制t细胞的持久性和抗肿瘤功效。我们发现,Fas变异的基因工程破坏了Fas受体在小鼠和人t细胞中作为显性负受体(dnr)结合FADD功能的能力,从而使细胞免于fasl诱导的凋亡。Fas dnr工程t细胞在ACT治疗后,在肿瘤内表现出增强的持久性,在用TCR或car修饰细胞治疗的实体和血液恶性肿瘤中,导致更好的癌症消退和总体生存率。尽管延长了寿命,Fas dnr工程t细胞没有发生异常克隆扩增,证明了这种方法的安全性。因此,细胞内在的“隔离”t细胞免受FasL的负面影响是一种潜在的通用策略,可以提高ACT在广泛的人类恶性肿瘤中的疗效。引用格式:Christopher A. k黎巴嫩诺夫。t细胞工程化克服肿瘤微环境中的死亡信号增强过继性癌症免疫治疗[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 PR05。
{"title":"Abstract PR05: T-cells engineered to overcome death signaling within the tumor microenvironment enhance adoptive cancer immunotherapy","authors":"C. Klebanoff","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-PR05","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-PR05","url":null,"abstract":"Across clinical trials, T-cell expansion and persistence following adoptive cell transfer (ACT) has correlated with superior patient outcomes. Herein, we undertook a pan-cancer analysis across human tumors to identify potentially actionable ligand/receptor pairs that might limit T-cell function and persistence following ACT. We discovered that FASLG, the gene encoding the apoptosis-inducing ligand FasL, is overexpressed within the majority of human tumor microenvironments. Further, we uncovered that Fas, the receptor for FasL, is highly expressed on patient-derived T-cells used for ACT. We hypothesized that a cognate Fas-FasL interaction within the tumor microenvironment might limit both T-cell persistence and antitumor efficacy. We discovered that genetic engineering of Fas variants impaired the ability to bind FADD function as dominant negative receptors (DNRs) in Fas-competent mouse and human T-cells, rescuing cells from FasL-induced apoptosis. Fas DNR-engineered T-cells exhibited enhanced persistence within tumors following ACT, resulting in superior cancer regression and overall survival in solid and hematologic malignancies treated with TCR or CAR-modified cells. Despite enhanced longevity, Fas DNR-engineered T-cells did not undergo aberrant clonal expansion, demonstrating the safety of this approach. Thus, cell-intrinsic “insulation” of T-cells from the negative influence of FasL is a potentially universal strategy to enhance ACT efficacy across a broad range of human malignancies. Citation Format: Christopher A. Klebanoff. T-cells engineered to overcome death signaling within the tumor microenvironment enhance adoptive cancer immunotherapy [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr PR05.","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"95 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":"123586697","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A027
Alexandra Croft, C. Brandt, Stephen M Burleigh, E. Chadwick, M. Chin, D. Toy, B. Donahue, Clay Patton, Stephen J. Goldfless, Brian J. Belmont, R. Salmon, G. Welstead, Blythe Sather, D. J. Huss
Background: Adoptive T-cell therapy with engineered T-cell receptors (eTCR) has demonstrated promising, yet modest clinical benefit to date. Key obstacles for this technology are 1) competition with the endogenous TCR for CD3 components that are required for surface expression of a functional TCR complex and 2) mispairing of endogenous and exogenous TCRα and TCRβ chains. Substituting murine TCR constant domains has been shown to enhance eTCR expression, but increases the risk of immunologic rejection. Knockout or knockdown of the endogenous TCR has also been shown to improve eTCR expression. Current eTCR delivery approaches use semi-randomly integrating lentivirus or retrovirus to generate an eTCR expressing T-cell product. An alternative approach is to combine TCR knockout with targeted integration of eTCRs into the T-cell receptor alpha constant (TRAC) locus. In this study, we evaluate newly discovered eTCRs specific for human papillomavirus (HPV) type 16 oncogenic protein E7, expressed via these various gene engineering approaches, to optimize engineered T-cell functionality. Methods: HPV E7-specific eTCRs were introduced into primary human T-cells by lentiviral transduction or a dual CRISPR-Cas9/AAV eTCR delivery platform for targeted insertion into the TRAC locus using homology directed repair (HDR). Comparisons were made among TCR sufficient and TCR knockouT-cells with gene delivery by lentivirus or HDR. Engineered T-cell function was assessed both in vitro and in vivo against HPV-16+ head and neck squamous cell carcinoma cell lines. Results: CRISPR/Cas9-mediated TRAC editing eliminated endogenous TCR expression in >85% of T-cells. The impact of TRAC editing on eTCR expression and engineered T-cell function was variable across multiple eTCRαβ sequences. With many eTCRs, TRAC editing in lentivirus-derived populations resulted in increased expression and improved cytokine and killing responses. Targeted insertion of these eTCRs into the TRAC locus by HDR increased engineered cell product homogeneity and enhanced function compared to lentiviral integration. However, there were also examples of eTCRs that were minimally impacted by TRAC locus engineering. Conclusions: Our results demonstrate that elimination of the endogenous TCR, alone or combined with targeted insertion at the TRAC locus, improves eTCR expression and engineered T-cell function for the majority of eTCRs tested. While infrequent, some eTCRs were less impacted by TRAC locus engineering, suggesting a sequence-specific property that enables these eTCRs to out-compete the endogenous TCR for CD3 componentry. The cause for this variable response to TRAC locus engineering is still being explored. Citation Format: Alexandra Croft, Cameron Brandt, Stephen Burleigh, Eric Chadwick, Melissa Chin, Dean Toy, Bailey Donahue, Clay Patton, Stephen Goldfless, Brian Belmont, Ruth Salmon, Grant Welstead, Blythe D. Sather, David J. Huss. Targeted insertion of an HPV-16 E7-specific engineered T-cell
背景:使用工程化t细胞受体(eTCR)的过继t细胞治疗已被证明是有希望的,但迄今为止的临床效果并不明显。该技术的主要障碍是1)与内源性TCR竞争CD3成分,这些成分是功能性TCR复合物表面表达所必需的;2)内源性和外源性TCRα和TCRβ链的错配。取代小鼠TCR恒定结构域已被证明可以增强eTCR表达,但增加了免疫排斥的风险。敲除或敲低内源性TCR也可改善eTCR的表达。目前的eTCR递送方法使用半随机整合慢病毒或逆转录病毒来产生表达t细胞产物的eTCR。另一种方法是将TCR基因敲除与etcr靶向整合到t细胞受体α常数(TRAC)位点相结合。在这项研究中,我们评估了新发现的针对人乳头瘤病毒(HPV) 16型致癌蛋白E7的特异性etcr,通过这些不同的基因工程方法表达,以优化工程化t细胞的功能。方法:将HPV e7特异性eTCR通过慢病毒转导或CRISPR-Cas9/AAV双重eTCR传递平台导入原代人t细胞,利用同源定向修复(homology directed repair, HDR)技术靶向插入TRAC位点。比较了慢病毒或HDR传递基因的TCR充足细胞和TCR敲除细胞。在体外和体内评估了工程t细胞对HPV-16+头颈部鳞状细胞癌细胞系的功能。结果:CRISPR/ cas9介导的TRAC编辑在>85%的t细胞中消除了内源性TCR表达。在多个eTCRαβ序列中,TRAC编辑对eTCR表达和工程t细胞功能的影响是不同的。对于许多etcr,在慢病毒衍生的群体中编辑TRAC导致表达增加、细胞因子和杀伤反应改善。与慢病毒整合相比,HDR将这些etcr靶向插入TRAC位点增加了工程细胞产物的均匀性和增强了功能。然而,也有受TRAC基因座工程影响最小的etcr的例子。结论:我们的研究结果表明,消除内源性TCR,单独或联合靶向插入TRAC位点,可以改善大多数eTCR的表达和工程t细胞功能。虽然不常见,但一些etcr受TRAC基因座工程的影响较小,这表明这些etcr具有序列特异性,能够与内源性TCR竞争CD3组分。这种对TRAC基因座工程的可变反应的原因仍在探索中。引文格式:Alexandra Croft, Cameron Brandt, Stephen Burleigh, Eric Chadwick, Melissa Chin, Dean Toy, Bailey Donahue, Clay Patton, Stephen Goldfless, Brian Belmont, Ruth Salmon, Grant Welstead, Blythe D. Sather, David J. Huss。将HPV-16 e7特异性工程化t细胞受体靶向插入TRAC位点[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A027。
{"title":"Abstract A027: Targeted insertion of an HPV-16 E7-specific engineered T-cell receptor into the TRAC locus","authors":"Alexandra Croft, C. Brandt, Stephen M Burleigh, E. Chadwick, M. Chin, D. Toy, B. Donahue, Clay Patton, Stephen J. Goldfless, Brian J. Belmont, R. Salmon, G. Welstead, Blythe Sather, D. J. Huss","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A027","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A027","url":null,"abstract":"Background: Adoptive T-cell therapy with engineered T-cell receptors (eTCR) has demonstrated promising, yet modest clinical benefit to date. Key obstacles for this technology are 1) competition with the endogenous TCR for CD3 components that are required for surface expression of a functional TCR complex and 2) mispairing of endogenous and exogenous TCRα and TCRβ chains. Substituting murine TCR constant domains has been shown to enhance eTCR expression, but increases the risk of immunologic rejection. Knockout or knockdown of the endogenous TCR has also been shown to improve eTCR expression. Current eTCR delivery approaches use semi-randomly integrating lentivirus or retrovirus to generate an eTCR expressing T-cell product. An alternative approach is to combine TCR knockout with targeted integration of eTCRs into the T-cell receptor alpha constant (TRAC) locus. In this study, we evaluate newly discovered eTCRs specific for human papillomavirus (HPV) type 16 oncogenic protein E7, expressed via these various gene engineering approaches, to optimize engineered T-cell functionality. Methods: HPV E7-specific eTCRs were introduced into primary human T-cells by lentiviral transduction or a dual CRISPR-Cas9/AAV eTCR delivery platform for targeted insertion into the TRAC locus using homology directed repair (HDR). Comparisons were made among TCR sufficient and TCR knockouT-cells with gene delivery by lentivirus or HDR. Engineered T-cell function was assessed both in vitro and in vivo against HPV-16+ head and neck squamous cell carcinoma cell lines. Results: CRISPR/Cas9-mediated TRAC editing eliminated endogenous TCR expression in >85% of T-cells. The impact of TRAC editing on eTCR expression and engineered T-cell function was variable across multiple eTCRαβ sequences. With many eTCRs, TRAC editing in lentivirus-derived populations resulted in increased expression and improved cytokine and killing responses. Targeted insertion of these eTCRs into the TRAC locus by HDR increased engineered cell product homogeneity and enhanced function compared to lentiviral integration. However, there were also examples of eTCRs that were minimally impacted by TRAC locus engineering. Conclusions: Our results demonstrate that elimination of the endogenous TCR, alone or combined with targeted insertion at the TRAC locus, improves eTCR expression and engineered T-cell function for the majority of eTCRs tested. While infrequent, some eTCRs were less impacted by TRAC locus engineering, suggesting a sequence-specific property that enables these eTCRs to out-compete the endogenous TCR for CD3 componentry. The cause for this variable response to TRAC locus engineering is still being explored. Citation Format: Alexandra Croft, Cameron Brandt, Stephen Burleigh, Eric Chadwick, Melissa Chin, Dean Toy, Bailey Donahue, Clay Patton, Stephen Goldfless, Brian Belmont, Ruth Salmon, Grant Welstead, Blythe D. Sather, David J. Huss. Targeted insertion of an HPV-16 E7-specific engineered T-cell","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"5 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":"129719461","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A028
L. Escribà-García, C. Álvarez-Fernández, A. Caballero, R. Julian, E. Hermann, J. Sierra, M. Hudecek, J. Briones
Peripheral T-cell lymphomas (PTCL) represent the most aggressive form among non-Hodgkin lymphomas with a very poor prognosis (5-year survival of 30%), demanding innovative novel treatment strategies. Adoptive immunotherapy with chimeric antigen receptor (CAR) T-cells has demonstrated its therapeutic potential in advanced B-cell hematologic malignancies. However, its application to PTCL remains a formidable challenge mainly due to a lack of truly tumor-specific antigens that are not expressed on normal T-cells. Anaplastic large T-cell lymphomas (ALCL) and several other subtypes of PTCL express CD30, which is also expressed by activated normal T-cells but no other healthy tissues. Indeed, brentuximab-vedotin, an anti-CD30 antibody-drug conjugate, has shown some clinical efficacy in PTCL and ALCL patients although duration of responses is short in the majority of cases. Here, we developed a refined CD30-CAR T-cell approach to target CD30+ PTCL as a potential novel therapeutic strategy. We selected a novel targeting domain that is unaffected by soluble CD30 protein (Nagata S et al., Clin Cancer Res 2002) to prevent blockade of the CD30-CAR. Moreover, we optimized the therapy by using memory stem T-cells (TSCM) to promote engraftment and persistence of CD30-CAR T-cells after transfer.TSCM were generated with CD3/CD28 costimulation in presence of IL-7, IL-15 and IL-21 (Alvarez C et al., J Transl Med 2016). On day 2 of culture, cells were transduced with a third-generation lentiviral vector encoding the CD30-41BBz-EGFRt CAR. The cell line Karpas 299 (ALCL) was used as tumor model. Cytotoxicity assay was performed at 24 hours and the tumor cell death was detected by luminiscence. Mice were injected with Karpas 299 tumor cells (2x10^6 cells/mice; iv) and were treated with CD30-CAR TSCM (1x10^7 cells/mice; iv) when the tumor was established (day 10). Mice were followed daily for survival. The presence of CD30-CAR TSCM in lymphoid organs and their expression of immune checkpoint molecules (i.e., TIM-3, LAG-3) were analyzed at the end of the in vivo study by flow cytometry. TSCM were the most prevalent T-cell subset at day 10 of culture (84±3.1% of total cells), and the CD30-CAR expression was 82.8±1.0% in CD4+ TSCM and 85.2±2.0% in CD8+ TSCM. Although CD30 protein was detected in a fraction of activated T-cells in culture, CD30-CAR TSCM could be expanded ex vivo (CD4+ CD30-CAR TSCM: 96.0±3.2 fold expansion; CD8+ CD30-CAR TSCM: 109.0±4.2 fold expansion). CD30-CAR TSCM conferred specific cytolytic activity against Karpas 299 cells in vitro (tumor cell death 5:1 (effector:target) ratio: 70.2±5.1% vs. 0% with untransduced TSCM; p 80% of detected CD4+ and CD8+ T-cells) were still present in lymphoid organs (bone marrow, spleen and lymph nodes) of surviving mice at 50 days after infusion. Tumor-bearing mice also showed CD30-CAR T-cells in their lymphoid organs (80% of total CD4+ and CD8+ T-cells) and in the tumor (67.5% of detected T-cells). Interestingly, a high
外周t细胞淋巴瘤(PTCL)是非霍奇金淋巴瘤中最具侵袭性的形式,预后非常差(5年生存率为30%),需要创新的新治疗策略。嵌合抗原受体(CAR) t细胞过继免疫疗法已证明其治疗晚期b细胞恶性血液病的潜力。然而,将其应用于PTCL仍然是一个巨大的挑战,主要是因为缺乏真正的肿瘤特异性抗原,这些抗原不在正常t细胞上表达。间变性大t细胞淋巴瘤(ALCL)和其他几种PTCL亚型表达CD30,活化的正常t细胞也表达CD30,但不表达其他健康组织。事实上,brentuximab-vedotin,一种抗cd30抗体-药物偶联物,在PTCL和ALCL患者中显示出一定的临床疗效,尽管大多数病例的反应持续时间较短。在这里,我们开发了一种改进的CD30- car - t细胞方法来靶向CD30+ PTCL,作为一种潜在的新型治疗策略。我们选择了一种不受可溶性CD30蛋白影响的新型靶向结构域(Nagata S等人,clincancer Res 2002)来阻止CD30- car的阻断。此外,我们通过使用记忆干细胞t细胞(TSCM)来优化治疗方法,以促进CD30-CAR - t细胞移植后的植入和持久性。在IL-7、IL-15和IL-21存在的情况下,通过CD3/CD28共刺激产生TSCM (Alvarez C et al., J Transl Med 2016)。在培养的第2天,用编码CD30-41BBz-EGFRt CAR的第三代慢病毒载体转导细胞。以Karpas 299 (ALCL)细胞系作为肿瘤模型。24h进行细胞毒性试验,荧光法检测肿瘤细胞死亡情况。小鼠注射Karpas 299肿瘤细胞(2 × 10^6个细胞/只;iv)和CD30-CAR TSCM处理(1 × 10^7个细胞/小鼠;Iv)肿瘤建立时(第10天)。每天跟踪小鼠存活。在体内研究结束时,用流式细胞术分析CD30-CAR TSCM在淋巴器官中的存在及其免疫检查点分子(即TIM-3、LAG-3)的表达。在培养第10天,TSCM是最常见的t细胞亚群(占总细胞的84±3.1%),CD30-CAR在CD4+ TSCM中的表达为82.8±1.0%,CD8+ TSCM中的表达为85.2±2.0%。虽然在培养的部分活化t细胞中检测到CD30蛋白,但CD30- car TSCM可以在体外扩增(CD4+ CD30- car TSCM:扩增96.0±3.2倍;CD8+ CD30-CAR TSCM: 109.0±4.2倍膨胀)。CD30-CAR TSCM对体外Karpas 299细胞具有特异性的细胞溶解活性(肿瘤细胞死亡5:1(效应器:靶标)比:70.2±5.1% vs.未转导的TSCM为0%;p注射50 d后,存活小鼠的淋巴器官(骨髓、脾脏和淋巴结)中仍有80%检测到的CD4+和CD8+ t细胞存在。荷瘤小鼠在其淋巴器官(占总CD4+和CD8+ t细胞的80%)和肿瘤(占检测到的t细胞的67.5%)中也显示出CD30-CAR - t细胞。有趣的是,在这些肿瘤浸润淋巴细胞(TILs)上发现了高表达的衰竭标志物TIM-3(74%±6.8%)和LAG-3(34%±9.7%)。总的来说,我们的数据表明,TSCM细胞可以用一种新的CD30- car有效地转导和体外扩增,并在体外和体内赋予抗CD30+ PTCL的有效抗肿瘤功效,使用免疫检查点阻断TIM-3或LAG-3的治疗可能会得到改善。我们的研究结果表明,通过CD30- car修饰t细胞的过继治疗,有可能改善CD30+ PTCL患者的预后。引文格式:Laura Escriba-Garcia, Carmen Alvarez-Fernandez, Ana Carolina Caballero, Rydzek Julian, Einsele Hermann, Jorge Sierra, Michael Hudecek, Javier Briones。表达优化的cd30特异性嵌合抗原受体(CAR)的记忆干细胞在体内有效地根除外周t细胞淋巴瘤[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 A028。
{"title":"Abstract A028: Memory stem T-cells expressing an optimized CD30-specific chimeric antigen receptor (CAR) efficiently eradicate peripheral T-cell lymphoma in vivo","authors":"L. Escribà-García, C. Álvarez-Fernández, A. Caballero, R. Julian, E. Hermann, J. Sierra, M. Hudecek, J. Briones","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A028","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A028","url":null,"abstract":"Peripheral T-cell lymphomas (PTCL) represent the most aggressive form among non-Hodgkin lymphomas with a very poor prognosis (5-year survival of 30%), demanding innovative novel treatment strategies. Adoptive immunotherapy with chimeric antigen receptor (CAR) T-cells has demonstrated its therapeutic potential in advanced B-cell hematologic malignancies. However, its application to PTCL remains a formidable challenge mainly due to a lack of truly tumor-specific antigens that are not expressed on normal T-cells. Anaplastic large T-cell lymphomas (ALCL) and several other subtypes of PTCL express CD30, which is also expressed by activated normal T-cells but no other healthy tissues. Indeed, brentuximab-vedotin, an anti-CD30 antibody-drug conjugate, has shown some clinical efficacy in PTCL and ALCL patients although duration of responses is short in the majority of cases. Here, we developed a refined CD30-CAR T-cell approach to target CD30+ PTCL as a potential novel therapeutic strategy. We selected a novel targeting domain that is unaffected by soluble CD30 protein (Nagata S et al., Clin Cancer Res 2002) to prevent blockade of the CD30-CAR. Moreover, we optimized the therapy by using memory stem T-cells (TSCM) to promote engraftment and persistence of CD30-CAR T-cells after transfer.TSCM were generated with CD3/CD28 costimulation in presence of IL-7, IL-15 and IL-21 (Alvarez C et al., J Transl Med 2016). On day 2 of culture, cells were transduced with a third-generation lentiviral vector encoding the CD30-41BBz-EGFRt CAR. The cell line Karpas 299 (ALCL) was used as tumor model. Cytotoxicity assay was performed at 24 hours and the tumor cell death was detected by luminiscence. Mice were injected with Karpas 299 tumor cells (2x10^6 cells/mice; iv) and were treated with CD30-CAR TSCM (1x10^7 cells/mice; iv) when the tumor was established (day 10). Mice were followed daily for survival. The presence of CD30-CAR TSCM in lymphoid organs and their expression of immune checkpoint molecules (i.e., TIM-3, LAG-3) were analyzed at the end of the in vivo study by flow cytometry. TSCM were the most prevalent T-cell subset at day 10 of culture (84±3.1% of total cells), and the CD30-CAR expression was 82.8±1.0% in CD4+ TSCM and 85.2±2.0% in CD8+ TSCM. Although CD30 protein was detected in a fraction of activated T-cells in culture, CD30-CAR TSCM could be expanded ex vivo (CD4+ CD30-CAR TSCM: 96.0±3.2 fold expansion; CD8+ CD30-CAR TSCM: 109.0±4.2 fold expansion). CD30-CAR TSCM conferred specific cytolytic activity against Karpas 299 cells in vitro (tumor cell death 5:1 (effector:target) ratio: 70.2±5.1% vs. 0% with untransduced TSCM; p 80% of detected CD4+ and CD8+ T-cells) were still present in lymphoid organs (bone marrow, spleen and lymph nodes) of surviving mice at 50 days after infusion. Tumor-bearing mice also showed CD30-CAR T-cells in their lymphoid organs (80% of total CD4+ and CD8+ T-cells) and in the tumor (67.5% of detected T-cells). Interestingly, a high","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"6 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":"129687118","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A044
Dy Lee, Rogier M. Reijmers, M. Honders, R. Hagedoorn, R. D. Jong, M. Kester, D. M. V. D. Steen, A. Ru, C. Kweekel, I. Jedema, H. Veelken, M. Heemskerk, P. Veelen, J. Falkenburg, M. Griffioen
The most frequent subtype of acute myeloid leukemia (AML) is defined by mutations in the nucleophosmin (NPM1) gene. Mutated NPM1 is an attractive target for immunotherapy, since it is an essential driver gene and 4 base pair frameshift insertions in exon 12 occur in 30-35% of AML, resulting in a novel C-terminal alternative reading frame of 11 amino acids. By searching in the HLA class I ligandome of primary AML, we identified multiple peptides derived from mutated NPM1. For one of these peptides, i.e., HLA-A*02:01-presented CLAVEEVSL, we searched for specific T-cells in AML patients and healthy individuals using peptide-MHC tetramers. Tetramer-positive CD8 T-cell clones were isolated and analyzed for reactivity against primary AML with mutated NPM1. From one selected clone with superior antitumor reactivity, we isolated the T-cell receptor (TCR) and demonstrated specific recognition and lysis of HLA-A*02:01-positive AML with mutated NPM1 in vitro after retroviral transfer to CD8 and CD4 T-cells. In vivo antitumor efficacy of TCR-transduced CD8 and CD4 T-cells was confirmed in immunodeficient mice engrafted with a human AML cell line expressing mutated NPM1. These data show that mutated NPM1-derived peptides are presented on AML and that CLAVEEVSL is a neoantigen that can be efficiently targeted on AML with mutated NPM1 by TCR gene transfer in a co-receptor independent fashion. Immunotherapy targeting mutated NPM1 may therefore contribute to treatment of AML. Citation Format: Dyantha I. van der Lee, Rogier M. Reijmers, M. Willy Honders, Renate S. Hagedoorn, Rob. M. de Jong, Michel G.D. Kester, Dirk M. van der Steen, Arnoud H. de Ru, Christiaan Kweekel, Inge Jedema, Hendrik Veelken, Mirjam M. Heemskerk, Peter A. van Veelen, J.H. Frederik Falkenburg, Marieke Griffioen. Mutated NPM1 as target for immunotherapy of acute myeloid leukemia [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 A044.
急性髓性白血病(AML)最常见的亚型是由核磷蛋白(NPM1)基因突变定义的。突变的NPM1是免疫治疗的一个有吸引力的靶标,因为它是一个重要的驱动基因,30-35%的AML患者在第12外显子上发生4个碱基对移码插入,导致新的c端11个氨基酸的替代阅读框。通过在原发性AML的HLA I类配体中搜索,我们发现了来自突变的NPM1的多个肽。对于其中一种肽,即HLA-A*02:01-present CLAVEEVSL,我们使用肽- mhc四聚体在AML患者和健康个体中寻找特异性t细胞。分离四聚体阳性CD8 t细胞克隆并分析其对NPM1突变的原发性AML的反应性。从一个具有优异抗肿瘤反应性的克隆中,我们分离了t细胞受体(TCR),并在体外证明了在将逆转录病毒转移到CD8和CD4 t细胞后,突变的NPM1对HLA-A*02:01阳性AML的特异性识别和裂解。在移植了表达突变NPM1的人AML细胞系的免疫缺陷小鼠中,证实了tcr转导的CD8和CD4 t细胞的体内抗肿瘤功效。这些数据表明,突变的NPM1衍生肽在AML上呈现,CLAVEEVSL是一种新抗原,可以通过TCR基因转移以一种独立于共受体的方式有效靶向突变的NPM1 AML。因此,针对突变的NPM1的免疫疗法可能有助于治疗AML。引用格式:Dyantha I. van der Lee, Rogier M. Reijmers, M. Willy Honders, Renate S. Hagedoorn, Rob。M. de Jong, Michel G.D. Kester, Dirk M. van der Steen, Arnoud H. de Ru, Christiaan Kweekel, Inge Jedema, Hendrik Veelken, Mirjam M. Heemskerk, Peter A. van Veelen, J.H. Frederik Falkenburg, Marieke Griffioen。突变的NPM1作为急性髓系白血病免疫治疗的靶点[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A044。
{"title":"Abstract A044: Mutated NPM1 as target for immunotherapy of acute myeloid leukemia","authors":"Dy Lee, Rogier M. Reijmers, M. Honders, R. Hagedoorn, R. D. Jong, M. Kester, D. M. V. D. Steen, A. Ru, C. Kweekel, I. Jedema, H. Veelken, M. Heemskerk, P. Veelen, J. Falkenburg, M. Griffioen","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A044","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A044","url":null,"abstract":"The most frequent subtype of acute myeloid leukemia (AML) is defined by mutations in the nucleophosmin (NPM1) gene. Mutated NPM1 is an attractive target for immunotherapy, since it is an essential driver gene and 4 base pair frameshift insertions in exon 12 occur in 30-35% of AML, resulting in a novel C-terminal alternative reading frame of 11 amino acids. By searching in the HLA class I ligandome of primary AML, we identified multiple peptides derived from mutated NPM1. For one of these peptides, i.e., HLA-A*02:01-presented CLAVEEVSL, we searched for specific T-cells in AML patients and healthy individuals using peptide-MHC tetramers. Tetramer-positive CD8 T-cell clones were isolated and analyzed for reactivity against primary AML with mutated NPM1. From one selected clone with superior antitumor reactivity, we isolated the T-cell receptor (TCR) and demonstrated specific recognition and lysis of HLA-A*02:01-positive AML with mutated NPM1 in vitro after retroviral transfer to CD8 and CD4 T-cells. In vivo antitumor efficacy of TCR-transduced CD8 and CD4 T-cells was confirmed in immunodeficient mice engrafted with a human AML cell line expressing mutated NPM1. These data show that mutated NPM1-derived peptides are presented on AML and that CLAVEEVSL is a neoantigen that can be efficiently targeted on AML with mutated NPM1 by TCR gene transfer in a co-receptor independent fashion. Immunotherapy targeting mutated NPM1 may therefore contribute to treatment of AML. Citation Format: Dyantha I. van der Lee, Rogier M. Reijmers, M. Willy Honders, Renate S. Hagedoorn, Rob. M. de Jong, Michel G.D. Kester, Dirk M. van der Steen, Arnoud H. de Ru, Christiaan Kweekel, Inge Jedema, Hendrik Veelken, Mirjam M. Heemskerk, Peter A. van Veelen, J.H. Frederik Falkenburg, Marieke Griffioen. Mutated NPM1 as target for immunotherapy of acute myeloid leukemia [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 A044.","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"80 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":"126786817","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A036
Cara-Lin Lonetree, Beau R. Webber, Miechaleen D. Diers, Walker S. Lahr, Mitchell G. Kluesner, Mark J. Osborn, Matthew J. Johnson, B. Moriarity
Chimeric antigen receptor (CAR)-engineered T-cells have mediated impressive outcomes in a subset of hematologic malignancies, yet this therapy remains highly personalized and largely ineffective against solid tumors. Genome editing strategies using targeted nucleases could overcome these limitations and have begun to enter clinical application. Multiplex editing strategies to develop off-the- shelf therapies are of high interest but remain limited by concerns of off-target effects and chromosomal translocations formed by simultaneous double-strand break (DSB) induction at multiple loci. The risk of genotoxic side effects is amplified when combining multiplex DSB induction with randomly integrating platforms for antigen-specific receptor delivery. An ideal strategy would allow for multigene disruption and targeted integration of antigen-specific receptors without introduction of multiple genomic DSBs. To this end, we evaluated the application of third- and fourth-generation Cas9 base editor technologies for gene disruption and integration in primary human T-cells. Chemically modified gRNAs and Cas9 base editor mRNA were delivered to stimulated T-cells by electroporation, followed by viral transduction for delivery of a DNA repair template as recombinant adeno-associated virus (rAAV). Base editing efficiencies were determined on the genomic level by PCR amplification, Sanger sequencing, and analysis of resultant traces using the EditR web app. Gene knock-out and knock-in efficiencies were analyzed on the protein level by flow cytometry. Through systematic reagent and dose optimization efforts, we achieved highly efficient C>T base conversion and consequent protein knockout at multiple therapeutically relevant loci including TRAC (KO = 83.6 ± 3.3%), PD-1 (KO = 78.6 ± 2.3%), and B2M (KO = 80 ± 1.8%). We observed that fourth-generation base editor (BE4) achieved consistently higher C>T conversion rates with reduced non-canonical editing (i.e., C>A/G) compared to third-generation base editor (BE3). Targeted disruption of splice acceptor (SA) and splice donor (SD) sites resulted in higher frequency of protein knockout vs. induction of premature stop codons at all loci examined. Importantly, while multiplex editing using Cas9 nuclease resulted in detectable translocations between the targeted sites, we were unable to detect these translocations using BE3 and BE4 as measured by PCR. Finally, we exploited the single-strand nickase function of the base editors in conjunction with rAAV delivery to achieve simultaneous targeted integration of a gene expression cassette at the AAVS1 safe harbor locus. Staphylococcus aureus Cas9 (SaCas9) was also employed as an orthogonal approach to AAVS1 targeting, and yielded higher rates of donor integration. Collectively, we demonstrate that Cas9 base editor technology can be utilized to mediate efficient, multiplex gene disruption and targeted gene integration in primary human T-cells without associated translocations. Th
嵌合抗原受体(CAR)工程t细胞在血液恶性肿瘤亚群中介导了令人印象深刻的结果,然而这种治疗仍然是高度个性化的,并且在很大程度上对实体肿瘤无效。使用靶向核酸酶的基因组编辑策略可以克服这些限制,并已开始进入临床应用。开发现成疗法的多重编辑策略受到高度关注,但仍然受到脱靶效应和多位点同时双链断裂(DSB)诱导形成的染色体易位的限制。当将多重DSB诱导与抗原特异性受体传递的随机整合平台相结合时,基因毒性副作用的风险被放大。理想的策略是在不引入多个基因组dsb的情况下,允许多基因破坏和抗原特异性受体的靶向整合。为此,我们评估了第三代和第四代Cas9碱基编辑器技术在原代人t细胞中基因破坏和整合的应用。化学修饰的gRNAs和Cas9碱基编辑器mRNA通过电穿孔传递到受刺激的t细胞,然后通过病毒转导以重组腺相关病毒(rAAV)的形式传递DNA修复模板。在基因组水平上,通过PCR扩增、Sanger测序和使用EditR web应用程序分析产生的痕量来确定碱基编辑效率。在蛋白质水平上,通过流式细胞术分析基因敲除和敲入效率。通过系统的试剂和剂量优化,我们实现了高效的C>T碱基转化和随后的蛋白敲除,包括TRAC (KO = 83.6±3.3%)、PD-1 (KO = 78.6±2.3%)和B2M (KO = 80±1.8%)。我们观察到,与第三代碱基编辑器(BE3)相比,第四代碱基编辑器(BE4)在减少非规范编辑(即C>A/G)的情况下实现了始终较高的C>T转化率。剪接受体(SA)和剪接供体(SD)位点的靶向破坏导致蛋白敲除频率高于诱导所有检测位点的过早终止密码子。重要的是,虽然使用Cas9核酸酶进行多重编辑导致目标位点之间可检测到的易位,但我们无法使用PCR测量的BE3和BE4检测到这些易位。最后,我们利用碱基编辑器的单链缺口酶功能与rAAV递送相结合,在AAVS1安全港位点同时实现了基因表达盒的靶向整合。金黄色葡萄球菌Cas9 (SaCas9)也被用作AAVS1靶向的正交方法,并产生更高的供体整合率。总的来说,我们证明了Cas9碱基编辑器技术可以在没有相关易位的情况下,在原代人t细胞中介导高效、多重基因破坏和靶向基因整合。这种简化的基因组工程方法可以广泛应用于开发安全有效的细胞疗法。引文格式:Cara Lonetree, Beau R. Webber, Miechaleen D. Diers, Walker S. Lahr, Mitchell Kluesner, Mark J. Osborn, Matthew J. Johnson, Branden S. Moriarity。基于Cas9碱基编辑器技术的多重人t细胞工程[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 A036。
{"title":"Abstract A036: Multiplex human T-cell engineering by Cas9 base editor technology","authors":"Cara-Lin Lonetree, Beau R. Webber, Miechaleen D. Diers, Walker S. Lahr, Mitchell G. Kluesner, Mark J. Osborn, Matthew J. Johnson, B. Moriarity","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A036","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A036","url":null,"abstract":"Chimeric antigen receptor (CAR)-engineered T-cells have mediated impressive outcomes in a subset of hematologic malignancies, yet this therapy remains highly personalized and largely ineffective against solid tumors. Genome editing strategies using targeted nucleases could overcome these limitations and have begun to enter clinical application. Multiplex editing strategies to develop off-the- shelf therapies are of high interest but remain limited by concerns of off-target effects and chromosomal translocations formed by simultaneous double-strand break (DSB) induction at multiple loci. The risk of genotoxic side effects is amplified when combining multiplex DSB induction with randomly integrating platforms for antigen-specific receptor delivery. An ideal strategy would allow for multigene disruption and targeted integration of antigen-specific receptors without introduction of multiple genomic DSBs. To this end, we evaluated the application of third- and fourth-generation Cas9 base editor technologies for gene disruption and integration in primary human T-cells. Chemically modified gRNAs and Cas9 base editor mRNA were delivered to stimulated T-cells by electroporation, followed by viral transduction for delivery of a DNA repair template as recombinant adeno-associated virus (rAAV). Base editing efficiencies were determined on the genomic level by PCR amplification, Sanger sequencing, and analysis of resultant traces using the EditR web app. Gene knock-out and knock-in efficiencies were analyzed on the protein level by flow cytometry. Through systematic reagent and dose optimization efforts, we achieved highly efficient C>T base conversion and consequent protein knockout at multiple therapeutically relevant loci including TRAC (KO = 83.6 ± 3.3%), PD-1 (KO = 78.6 ± 2.3%), and B2M (KO = 80 ± 1.8%). We observed that fourth-generation base editor (BE4) achieved consistently higher C>T conversion rates with reduced non-canonical editing (i.e., C>A/G) compared to third-generation base editor (BE3). Targeted disruption of splice acceptor (SA) and splice donor (SD) sites resulted in higher frequency of protein knockout vs. induction of premature stop codons at all loci examined. Importantly, while multiplex editing using Cas9 nuclease resulted in detectable translocations between the targeted sites, we were unable to detect these translocations using BE3 and BE4 as measured by PCR. Finally, we exploited the single-strand nickase function of the base editors in conjunction with rAAV delivery to achieve simultaneous targeted integration of a gene expression cassette at the AAVS1 safe harbor locus. Staphylococcus aureus Cas9 (SaCas9) was also employed as an orthogonal approach to AAVS1 targeting, and yielded higher rates of donor integration. Collectively, we demonstrate that Cas9 base editor technology can be utilized to mediate efficient, multiplex gene disruption and targeted gene integration in primary human T-cells without associated translocations. Th","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"285 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":"116103498","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-IA12
J. Kochenderfer
Chimeric antigen receptors (CARs) are fusion proteins that incorporate antigen-binding domains and T-cell signaling domains. The first CAR T cells to be shown to be clinically effective targeted the B-cell antigen CD19. Anti-CD19 CAR T cells have been effective against all major types of B-cell lymphomas, and anti-CD19 CAR T cells have now obtained Food and Drug Administration (FDA) approval for treatment of the most common type of lymphoma, diffuse large B-cell lymphoma. This approval was based on clinical results that showed an approximately 50% complete remission rate of anti-CD19 CAR T-cell therapy for diffuse large B-cell lymphoma. These responses have been durable. Eleven of twelve patients who obtained complete remissions on a phase 1 trial of anti-CD19 CAR T cells are still progression-free 13 to 36 months after treatment (Kochenderfer et al. Journal of Clinical Oncology, 2017). Even more durable remissions have occurred in a small number of patients in an early cohort of anti-CD19 CAR therapy. Four patients with remissions of 38 to 56 months will be presented. These patients are particularly interesting because 3 of the 4 patients with long remissions have had recovery of normal B cells while the complete remissions have continued. This result shows that remissions of diffuse large B-cell lymphoma can continue after disappearance of a functional anti-CD19 CAR T-cell response. Beyond CD19, active CAR T-cell therapy for multiple myeloma has been developed by targeting B-cell maturation antigen (BCMA). Anti-BCMA CAR T cells have undergone rapid clinical development since first being reported in 2013 (Carpenter et al. Clinical Cancer Research, 2013). Clinical results of the first-in-humans clinical trial of anti-BCMA CARs (Brudno et al. Journal of Clinical Oncology, 2018) will be presented, and future plans for multiple myeloma CAR T-cell therapies will be discussed. Citation Format: James N. Kochenderfer. CAR T-cell therapy for lymphoma and multiple myeloma [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 IA12.
嵌合抗原受体(CARs)是结合抗原结合域和t细胞信号域的融合蛋白。第一个CAR - T细胞被证明是临床有效的靶向b细胞抗原CD19。抗cd19 CAR - T细胞对所有主要类型的b细胞淋巴瘤都有效,目前,抗cd19 CAR - T细胞已获得美国食品和药物管理局(FDA)批准,可用于治疗最常见的淋巴瘤——弥漫性大b细胞淋巴瘤。该批准是基于临床结果,显示抗cd19 CAR - t细胞治疗弥漫性大b细胞淋巴瘤的完全缓解率约为50%。这些反应是持久的。在抗cd19 CAR - T细胞1期试验中获得完全缓解的12名患者中,有11名在治疗后13至36个月仍无进展(Kochenderfer等)。临床肿瘤学杂志,2017)。在抗cd19 CAR治疗的早期队列中,在少数患者中出现了更持久的缓解。4例患者的缓解期为38至56个月。这些患者特别有趣因为4个长期缓解的患者中有3个恢复了正常的B细胞而完全缓解仍在继续。这一结果表明,弥漫性大b细胞淋巴瘤的缓解可以在功能性抗cd19 CAR - t细胞反应消失后继续。除了CD19,针对b细胞成熟抗原(BCMA)的活性CAR - t细胞治疗多发性骨髓瘤已经被开发出来。Anti-BCMA CAR - T细胞自2013年首次报道以来,经历了快速的临床发展(Carpenter等)。临床癌症研究,2013)。抗bcma CARs首次人体临床试验的临床结果(Brudno et al.)。临床肿瘤学杂志,2018),并将讨论多发性骨髓瘤CAR - t细胞治疗的未来计划。引用格式:James N. Kochenderfer。CAR - t细胞治疗淋巴瘤和多发性骨髓瘤[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1 - 12。
{"title":"Abstract IA12: CAR T-cell therapy for lymphoma and multiple myeloma","authors":"J. Kochenderfer","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-IA12","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-IA12","url":null,"abstract":"Chimeric antigen receptors (CARs) are fusion proteins that incorporate antigen-binding domains and T-cell signaling domains. The first CAR T cells to be shown to be clinically effective targeted the B-cell antigen CD19. Anti-CD19 CAR T cells have been effective against all major types of B-cell lymphomas, and anti-CD19 CAR T cells have now obtained Food and Drug Administration (FDA) approval for treatment of the most common type of lymphoma, diffuse large B-cell lymphoma. This approval was based on clinical results that showed an approximately 50% complete remission rate of anti-CD19 CAR T-cell therapy for diffuse large B-cell lymphoma. These responses have been durable. Eleven of twelve patients who obtained complete remissions on a phase 1 trial of anti-CD19 CAR T cells are still progression-free 13 to 36 months after treatment (Kochenderfer et al. Journal of Clinical Oncology, 2017). Even more durable remissions have occurred in a small number of patients in an early cohort of anti-CD19 CAR therapy. Four patients with remissions of 38 to 56 months will be presented. These patients are particularly interesting because 3 of the 4 patients with long remissions have had recovery of normal B cells while the complete remissions have continued. This result shows that remissions of diffuse large B-cell lymphoma can continue after disappearance of a functional anti-CD19 CAR T-cell response. Beyond CD19, active CAR T-cell therapy for multiple myeloma has been developed by targeting B-cell maturation antigen (BCMA). Anti-BCMA CAR T cells have undergone rapid clinical development since first being reported in 2013 (Carpenter et al. Clinical Cancer Research, 2013). Clinical results of the first-in-humans clinical trial of anti-BCMA CARs (Brudno et al. Journal of Clinical Oncology, 2018) will be presented, and future plans for multiple myeloma CAR T-cell therapies will be discussed. Citation Format: James N. Kochenderfer. CAR T-cell therapy for lymphoma and multiple myeloma [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 IA12.","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"365 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":"133384388","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A040
P. Rennert, Fay J. Dufort, Lihe Su, Lan Wu, A. Birt, C. Ambrose, R. Lobb
Introduction: CAR T-cells targeting CD19 (CAR19s) can eradicate B cell leukemias and lymphomas. The effectiveness of CAR19s is linked to their robust expansion properties but also their long-term persistence. Persistence is maintained by normal CD19+ B cells: a non-tumor dependent, self-renewing source of antigen. In this manner, CAR19s are quite unique. We have re-engineered CAR19s to secrete a wide variety of retargeting fusion proteins (FPs) by encoding expression cassettes downstream of the CAR sequence in lentiviral vectors. By hijacking CAR19s, we utilize their inherent persistence properties. By designing multispecific FP, we directly counter the clinically critical issues of tumor heterogeneity and antigen loss. Experimental Procedures: A lentiviral vector with an MCSV promoter was used to express the CAR19 construct and FPs. FPs and multispecific-FPs were designed to encode the extracellular domain of the CD19 protein, followed by one or two scFv sequences, separated from the CAR sequence by a P2A cleavage site, a design termed IMPACTtm (Integrated Modular Proteins for Adoptive Cell Therapy). The FPs therefore consist of the CD19 extracellular domain linked to one or more scFvs. The FPs redirect CAR19 T-cell cytotoxic activity to any tumor antigen of interest by coating that antigen with CD19 via the scFv. Further, multiple antigens can be coated with CD19 by encoding multiple scFv in the FP. Hijacked CAR19s therefore serve as a platform for targeting diverse antigens. Results: Here we describe one example in detail, focusing on Her2+ solid tumors. The CD19/anti-Her2 FP was highly potent in cytotoxicity assays targeting Her2+/CD19- solid tumor cell lines. The concentration of fusion protein required to reduce tumor cell number by 50% was 10 pM (0.7 ng/ml). Primary donor T-cells transduced with the CAR19 - CD19/anti-Her2 FP lentiviral vector secreted > 20 ng/ml of FP in cell culture. Cytotoxic activity of this redirected CAR19 against Her2+ SKOV3 tumor cells was demonstrated in vitro and in vivo. CD19-mediated persistence was demonstrated in serial restimulation assays. A bispecific FP containing CD19 linked to anti-Her2 and anti-EGFR scFv had specific activity against both antigens with a potency of 0.75 pM. For each antigen, the potent cytotoxicity was specifically mediated by the secreted fusion protein. Additional program examples of multispecific targeting for diverse hematologic and solid tumor types will be shown. Conclusions: The IMPACT platform addresses critical issues in cell therapy including CAR persistence, antigen escape and antigen heterogeneity, and provides important solutions for treating both hematologic and solid tumors. The potency of redirected cytotoxicity supports clinical development of CAR19/IMPACT programs, four of which are now ready for IND enabling studies. Citation Format: Paul Rennert, Fay Dufort, Lihe Su, Lan Wu, Alyssa Birt, Christine Ambrose, Roy Lobb. Hijacking CAR19 T-cells for use in targeting divers
导语:靶向CD19的CAR - t细胞(car19)可以根除B细胞白血病和淋巴瘤。car19的有效性与其强大的膨胀特性有关,但也与其长期持久性有关。持续性由正常CD19+ B细胞维持:一种非肿瘤依赖性、自我更新的抗原来源。在这方面,car19是非常独特的。我们重新设计了car19,通过在慢病毒载体中编码CAR序列下游的表达磁带,使其分泌多种重靶向融合蛋白(FPs)。通过劫持car19,我们利用了它们固有的持久性。通过设计多特异性FP,我们直接解决了肿瘤异质性和抗原丢失的临床关键问题。实验步骤:使用带MCSV启动子的慢病毒载体表达CAR19结构和FPs。FPs和多特异性FPs被设计用于编码CD19蛋白的细胞外结构域,随后是一个或两个scFv序列,通过P2A切割位点与CAR序列分离,这种设计称为IMPACTtm (Integrated Modular Proteins for Adoptive Cell Therapy)。因此,FPs由连接一个或多个scFvs的CD19细胞外结构域组成。FPs通过scFv将CD19包裹在任何感兴趣的肿瘤抗原上,从而将CAR19 t细胞的细胞毒性活性转移到该抗原上。此外,通过在FP中编码多个scFv,多种抗原可以被CD19包裹。因此,被劫持的car19作为靶向多种抗原的平台。结果:这里我们详细描述一个例子,重点是Her2+实体瘤。CD19/anti-Her2 FP在针对Her2+/CD19-实体瘤细胞系的细胞毒实验中表现出很强的效力。使肿瘤细胞数量减少50%所需的融合蛋白浓度为10 pM (0.7 ng/ml)。用CAR19 - CD19/anti-Her2 FP慢病毒载体转导的原代供体t细胞在细胞培养中分泌20 ng/ml的FP。在体外和体内均证实了这种重定向的CAR19对Her2+ SKOV3肿瘤细胞的细胞毒活性。cd19介导的持久性在一系列再刺激试验中得到证实。含有CD19的双特异性FP与抗her2和抗egfr连接,对这两种抗原具有特异性活性,效价为0.75 pM。对于每种抗原,有效的细胞毒性是由分泌的融合蛋白特异性介导的。将展示针对不同血液学和实体肿瘤类型的多特异性靶向的其他程序示例。结论:IMPACT平台解决了细胞治疗中的关键问题,包括CAR持久性、抗原逃逸和抗原异质性,为血液和实体肿瘤的治疗提供了重要的解决方案。重定向细胞毒性的效力支持CAR19/IMPACT项目的临床开发,其中四个项目现已准备好进行IND研究。引用格式:Paul Rennert, Fay Dufort, Lihe Su, Lan Wu, Alyssa Birt, Christine Ambrose, Roy Lobb。劫持CAR19 t细胞用于靶向多种造血和实体肿瘤[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 A040。
{"title":"Abstract A040: Hijacking CAR19 T-cells for use in targeting diverse hematopoietic and solid tumors","authors":"P. Rennert, Fay J. Dufort, Lihe Su, Lan Wu, A. Birt, C. Ambrose, R. Lobb","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A040","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A040","url":null,"abstract":"Introduction: CAR T-cells targeting CD19 (CAR19s) can eradicate B cell leukemias and lymphomas. The effectiveness of CAR19s is linked to their robust expansion properties but also their long-term persistence. Persistence is maintained by normal CD19+ B cells: a non-tumor dependent, self-renewing source of antigen. In this manner, CAR19s are quite unique. We have re-engineered CAR19s to secrete a wide variety of retargeting fusion proteins (FPs) by encoding expression cassettes downstream of the CAR sequence in lentiviral vectors. By hijacking CAR19s, we utilize their inherent persistence properties. By designing multispecific FP, we directly counter the clinically critical issues of tumor heterogeneity and antigen loss. Experimental Procedures: A lentiviral vector with an MCSV promoter was used to express the CAR19 construct and FPs. FPs and multispecific-FPs were designed to encode the extracellular domain of the CD19 protein, followed by one or two scFv sequences, separated from the CAR sequence by a P2A cleavage site, a design termed IMPACTtm (Integrated Modular Proteins for Adoptive Cell Therapy). The FPs therefore consist of the CD19 extracellular domain linked to one or more scFvs. The FPs redirect CAR19 T-cell cytotoxic activity to any tumor antigen of interest by coating that antigen with CD19 via the scFv. Further, multiple antigens can be coated with CD19 by encoding multiple scFv in the FP. Hijacked CAR19s therefore serve as a platform for targeting diverse antigens. Results: Here we describe one example in detail, focusing on Her2+ solid tumors. The CD19/anti-Her2 FP was highly potent in cytotoxicity assays targeting Her2+/CD19- solid tumor cell lines. The concentration of fusion protein required to reduce tumor cell number by 50% was 10 pM (0.7 ng/ml). Primary donor T-cells transduced with the CAR19 - CD19/anti-Her2 FP lentiviral vector secreted > 20 ng/ml of FP in cell culture. Cytotoxic activity of this redirected CAR19 against Her2+ SKOV3 tumor cells was demonstrated in vitro and in vivo. CD19-mediated persistence was demonstrated in serial restimulation assays. A bispecific FP containing CD19 linked to anti-Her2 and anti-EGFR scFv had specific activity against both antigens with a potency of 0.75 pM. For each antigen, the potent cytotoxicity was specifically mediated by the secreted fusion protein. Additional program examples of multispecific targeting for diverse hematologic and solid tumor types will be shown. Conclusions: The IMPACT platform addresses critical issues in cell therapy including CAR persistence, antigen escape and antigen heterogeneity, and provides important solutions for treating both hematologic and solid tumors. The potency of redirected cytotoxicity supports clinical development of CAR19/IMPACT programs, four of which are now ready for IND enabling studies. Citation Format: Paul Rennert, Fay Dufort, Lihe Su, Lan Wu, Alyssa Birt, Christine Ambrose, Roy Lobb. Hijacking CAR19 T-cells for use in targeting divers","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"53 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":"127271288","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A029
Laura Evgin, Amanda L. Huff, Phonphimon Wongthida, Jill Thompson, T. Kottke, J. Sampson, L. Perez, R. Vile
Although the chimeric antigen receptor (CAR) T-cell platform has experienced clinical success in patients with hematologic malignancies, CAR T-cells specific to solid tumor targets have met with more limited efficacy. The highly inflammatory nature of oncolytic viruses and their ability to remodel the tumor microenvironment suggested to us that they would provide a complementary mechanism of action to both recruit and potentiate the functionality of CAR T-cells. VSVmIFNβ injection into B16EGFRvIII tumors increased the expression of chemokines such as CXCL10 and CCL5, which we would expect to recruit CXCR3+and CCR5+ EGFRvIII specific murine CAR T-cells. However, we did not observe an increase in overall survival or tumor control using the combination strategy compared to monotherapy with CAR T-cells. We recovered fewer viable CD8+ CAR T-cells from tumors injected with VSVmIFNβ early after adoptive transfer, and observed a similar reduction in the number of CD8+ CAR T-cells which persisted long term in the blood. We have shown that type I interferon increases the expression of the CAR from the retroviral LTR and in turn sensitizes these cells to tonic signaling mediated exhaustion and apoptosis. Correspondingly, CAR T-cells prepared from IFNAR KO T-cells were protected from the deleterious effect of type I IFN in vivo. We are currently investigating strategies to overcome the interference between these two modalities and to uncouple the regulation of the expression of the CAR from type I IFN signaling. Citation Format: Laura Evgin, Amanda L. Huff, Phonphimon Wongthida, Jill Thompson, Timothy Kottke, John Sampson, Luis Sanchez Perez, Richard Vile. Unexpected antagonism between oncolytic virus derived type I interferon and EGFRvIII 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 A029.
尽管嵌合抗原受体(CAR) t细胞平台在血液系统恶性肿瘤患者中取得了临床成功,但针对实体肿瘤靶点的CAR - t细胞的疗效有限。溶瘤病毒的高度炎症性质及其重塑肿瘤微环境的能力向我们表明,它们将提供一种互补的作用机制,以招募和增强CAR - t细胞的功能。将VSVmIFNβ注射到B16EGFRvIII肿瘤中增加了趋化因子如CXCL10和CCL5的表达,我们预计这些趋化因子会募集CXCR3+和CCR5+ EGFRvIII特异性小鼠CAR - t细胞。然而,与CAR - t细胞单药治疗相比,我们没有观察到使用联合策略的总生存期或肿瘤控制的增加。我们在过继转移后早期从注射了VSVmIFNβ的肿瘤中回收了较少的活CD8+ CAR - t细胞,并观察到在血液中长期存在的CD8+ CAR - t细胞数量的类似减少。我们已经证明,I型干扰素增加了来自逆转录病毒LTR的CAR的表达,并反过来使这些细胞对滋补信号介导的衰竭和凋亡敏感。相应地,IFNAR KO t细胞制备的CAR - t细胞在体内可以免受I型IFN的有害作用。我们目前正在研究克服这两种模式之间干扰的策略,并从I型IFN信号中解耦CAR表达的调节。引文格式:Laura Evgin, Amanda L. Huff, Phonphimon Wongthida, Jill Thompson, Timothy Kottke, John Sampson, Luis Sanchez Perez, Richard Vile。溶瘤病毒衍生的I型干扰素与EGFRvIII CAR - t细胞之间的意外拮抗作用[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 A029。
{"title":"Abstract A029: Unexpected antagonism between oncolytic virus derived type I interferon and EGFRvIII CAR T-cells","authors":"Laura Evgin, Amanda L. Huff, Phonphimon Wongthida, Jill Thompson, T. Kottke, J. Sampson, L. Perez, R. Vile","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A029","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A029","url":null,"abstract":"Although the chimeric antigen receptor (CAR) T-cell platform has experienced clinical success in patients with hematologic malignancies, CAR T-cells specific to solid tumor targets have met with more limited efficacy. The highly inflammatory nature of oncolytic viruses and their ability to remodel the tumor microenvironment suggested to us that they would provide a complementary mechanism of action to both recruit and potentiate the functionality of CAR T-cells. VSVmIFNβ injection into B16EGFRvIII tumors increased the expression of chemokines such as CXCL10 and CCL5, which we would expect to recruit CXCR3+and CCR5+ EGFRvIII specific murine CAR T-cells. However, we did not observe an increase in overall survival or tumor control using the combination strategy compared to monotherapy with CAR T-cells. We recovered fewer viable CD8+ CAR T-cells from tumors injected with VSVmIFNβ early after adoptive transfer, and observed a similar reduction in the number of CD8+ CAR T-cells which persisted long term in the blood. We have shown that type I interferon increases the expression of the CAR from the retroviral LTR and in turn sensitizes these cells to tonic signaling mediated exhaustion and apoptosis. Correspondingly, CAR T-cells prepared from IFNAR KO T-cells were protected from the deleterious effect of type I IFN in vivo. We are currently investigating strategies to overcome the interference between these two modalities and to uncouple the regulation of the expression of the CAR from type I IFN signaling. Citation Format: Laura Evgin, Amanda L. Huff, Phonphimon Wongthida, Jill Thompson, Timothy Kottke, John Sampson, Luis Sanchez Perez, Richard Vile. Unexpected antagonism between oncolytic virus derived type I interferon and EGFRvIII 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 A029.","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"1 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":"116030753","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A042
P. Schmidt, Matthias Bozza, D. Jaeger, R. Harbottle
Adoptive immunotherapy is one of the most encouraging therapeutic strategies for the treatment of a range of cancers. A particularly promising avenue of research is the functional introduction of chimeric antigen receptors (CARs) into naive human T-cells for autologous immunotherapy. Currently, the genetic engineering of these cells is achieved through the use of integrating vector systems such as lentiviruses or the sleeping beauty transposon system, which present a potential risk of genotoxicity associated with their random genomic integration. We have invented a novel DNA vector platform for the safe and efficient generation of genetically engineered T-cells for human immunotherapy. This DNA vector system contains no viral components and comprises only clinically approved sequences; it does not integrate into the target-cell’s genome but it can replicate autonomously and extrachromosomally in the nuclei of dividing human primary cells. These DNA vectors offer several advantages over currently used vector systems; they are not subject to commercial licenses, they are cheaper and easier to produce, and they can more quickly genetically modify human cells without the inherent risk of integrative mutagenesis. In preclinical experiments we have successfully generated genetically engineered human T-cells that sustain the expression of a reporter gene for over a month at persistently high levels without decline. We have also successfully modified these cells with a range of transgenic CAR receptors against several known cancer cell epitopes and we have demonstrated their viability and capability in the targeted killing of these human cancer cells. We showed that CAR-T-cells generated with our technology killed more efficiently target cells when compared to T-cells engineered with current state-of-the-art integrative lentivirus. The expression of functional CARs was detected over a period of two weeks of administration in culture and the anticancer activity of our DNA-CAR-T-cells was evaluated in vivo using xenotransplanted cell lines in immunodeficient mice. We are currently performing analyses in order to determine the molecular behavior of the vector in the cells and its impact on cellular viability. Furthermore, we are developing a protocol for large scale electroporation in order to manufacture a clinical grade CAR-T DNA product. We believe that this novel DNA vector system provides a unique and innovative approach to this exciting therapeutic strategy for cancer therapy. We estimate that this novel methodology will provide a simpler method of CAR T-cell manufacturing, resulting in a 10-fold reduction in the cost of the CAR-T product. Citation Format: Patrick Schmidt, Matthias Bozza, Dirk Jaeger, Richard Harbottle. A novel nonviral, nonintegrative DNA vector system for T-cell engineering [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 201
过继免疫疗法是治疗一系列癌症的最令人鼓舞的治疗策略之一。一个特别有前途的研究途径是将嵌合抗原受体(CARs)引入幼稚的人t细胞中进行自体免疫治疗。目前,这些细胞的基因工程是通过使用整合载体系统(如慢病毒或睡美人转座子系统)来实现的,这些系统存在与随机基因组整合相关的潜在遗传毒性风险。我们已经发明了一种新的DNA载体平台,用于安全有效地生成用于人类免疫治疗的基因工程t细胞。该DNA载体系统不含病毒成分,只包含临床批准的序列;它不会整合到目标细胞的基因组中,但它可以在分裂的人类原代细胞的细胞核中自主复制和染色体外复制。与目前使用的载体系统相比,这些DNA载体提供了几个优势;它们不受商业许可的约束,它们更便宜,更容易生产,而且它们可以更快地对人类细胞进行基因改造,而不会产生整合突变的固有风险。在临床前实验中,我们已经成功地产生了基因工程的人类t细胞,这种细胞可以持续一个多月的高水平表达报告基因而不会下降。我们还成功地用一系列针对几种已知癌细胞表位的转基因CAR受体修饰了这些细胞,并证明了它们在靶向杀死这些人类癌细胞方面的生存能力和能力。我们发现,与使用当前最先进的整合慢病毒设计的t细胞相比,用我们的技术生成的car -t细胞能更有效地杀死靶细胞。在培养中检测功能性car - t细胞的表达,并在免疫缺陷小鼠体内使用异种移植细胞系评估我们的dna - car - t细胞的抗癌活性。我们目前正在进行分析,以确定载体在细胞中的分子行为及其对细胞活力的影响。此外,我们正在开发一种大规模电穿孔的方案,以制造临床级CAR-T DNA产品。我们相信这种新的DNA载体系统为这种令人兴奋的癌症治疗策略提供了一种独特而创新的方法。我们估计,这种新方法将提供一种更简单的CAR- t细胞制造方法,从而使CAR- t产品的成本降低10倍。引用格式:Patrick Schmidt, Matthias Bozza, Dirk Jaeger, Richard Harbottle。一种用于t细胞工程的新型非病毒、非整合DNA载体系统[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 A042。
{"title":"Abstract A042: A novel nonviral, nonintegrative DNA vector system for T-cell engineering","authors":"P. Schmidt, Matthias Bozza, D. Jaeger, R. Harbottle","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A042","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A042","url":null,"abstract":"Adoptive immunotherapy is one of the most encouraging therapeutic strategies for the treatment of a range of cancers. A particularly promising avenue of research is the functional introduction of chimeric antigen receptors (CARs) into naive human T-cells for autologous immunotherapy. Currently, the genetic engineering of these cells is achieved through the use of integrating vector systems such as lentiviruses or the sleeping beauty transposon system, which present a potential risk of genotoxicity associated with their random genomic integration. We have invented a novel DNA vector platform for the safe and efficient generation of genetically engineered T-cells for human immunotherapy. This DNA vector system contains no viral components and comprises only clinically approved sequences; it does not integrate into the target-cell’s genome but it can replicate autonomously and extrachromosomally in the nuclei of dividing human primary cells. These DNA vectors offer several advantages over currently used vector systems; they are not subject to commercial licenses, they are cheaper and easier to produce, and they can more quickly genetically modify human cells without the inherent risk of integrative mutagenesis. In preclinical experiments we have successfully generated genetically engineered human T-cells that sustain the expression of a reporter gene for over a month at persistently high levels without decline. We have also successfully modified these cells with a range of transgenic CAR receptors against several known cancer cell epitopes and we have demonstrated their viability and capability in the targeted killing of these human cancer cells. We showed that CAR-T-cells generated with our technology killed more efficiently target cells when compared to T-cells engineered with current state-of-the-art integrative lentivirus. The expression of functional CARs was detected over a period of two weeks of administration in culture and the anticancer activity of our DNA-CAR-T-cells was evaluated in vivo using xenotransplanted cell lines in immunodeficient mice. We are currently performing analyses in order to determine the molecular behavior of the vector in the cells and its impact on cellular viability. Furthermore, we are developing a protocol for large scale electroporation in order to manufacture a clinical grade CAR-T DNA product. We believe that this novel DNA vector system provides a unique and innovative approach to this exciting therapeutic strategy for cancer therapy. We estimate that this novel methodology will provide a simpler method of CAR T-cell manufacturing, resulting in a 10-fold reduction in the cost of the CAR-T product. Citation Format: Patrick Schmidt, Matthias Bozza, Dirk Jaeger, Richard Harbottle. A novel nonviral, nonintegrative DNA vector system for T-cell engineering [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 201","PeriodicalId":254712,"journal":{"name":"Genetically Engineered T-cells","volume":"86 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":"124738532","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}