首页 > 最新文献

Molecular Therapy最新文献

英文 中文
AAV-mediated Stambp gene replacement therapy rescues neurological defects in a mouse model of microcephaly-capillary malformation syndrome. AAV介导的Stambp基因替代疗法可挽救小头畸形-毛细血管畸形综合征小鼠模型的神经缺陷。
IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-08-22 DOI: 10.1016/j.ymthe.2024.08.017
Meixin Hu, Jun Li, Jingxin Deng, Chunxue Liu, Yingying Liu, Huiping Li, Weijun Feng, Xiu Xu

The microcephaly-capillary malformation (MIC-CAP) syndrome is a life-threatening disease caused by biallelic mutations of the STAMBP gene, which encodes an endosomal deubiquitinating enzyme. To establish a suitable preclinical animal model for clinical therapeutic practice, we generated a central nervous system (CNS)-specific Stambp knockout mouse model (Stambp Sox1-cKO) that phenocopies Stambp null mice including progressive microcephaly, postnatal growth retardation and complete penetrance of preweaning death. In this MIC-CAP syndrome mouse model, early-onset neuronal death occurs specifically in the hippocampus and cortex, accompanied by aggregation of ubiquitinated proteins, and massive neuroinflammation. Importantly, neonatal AAV9-mediated gene supplementation of Stambp in the brain could significantly improve neurological defects, sustain growth, and prolong the lifespan of StambpSox1-cKO mice. Together, our findings reveal a central role of brain defects in the pathogenesis of STAMBP deficiency and provide preclinical evidence that postnatal gene replacement is an effective approach to cure the disease.

小头畸形-毛细血管畸形(MIC-CAP)综合征是一种威胁生命的疾病,由 STAMBP 基因的双倍突变引起,STAMBP 基因编码一种内体去泛素化酶。为了建立一个适用于临床治疗的临床前动物模型,我们建立了一个中枢神经系统(CNS)特异性 Stambp 基因敲除小鼠模型(StambpSox1-cKO),该模型可复制 Stambp 基因缺失小鼠的表型,包括进行性小头畸形、出生后生长迟缓和断奶前死亡的完全穿透性。在这种 MIC-CAP 综合征小鼠模型中,早发性神经元死亡特别发生在海马和皮层,同时伴有泛素化蛋白的聚集和大规模神经炎症。重要的是,新生儿期 AAV9 介导的脑内 Stambp 基因补充能显著改善 StambpSox1-cKO 小鼠的神经系统缺陷、维持生长并延长寿命。总之,我们的研究结果揭示了大脑缺陷在 STAMBP 缺乏症发病机制中的核心作用,并提供了临床前证据,证明产后基因替代是治疗该病的有效方法。
{"title":"AAV-mediated Stambp gene replacement therapy rescues neurological defects in a mouse model of microcephaly-capillary malformation syndrome.","authors":"Meixin Hu, Jun Li, Jingxin Deng, Chunxue Liu, Yingying Liu, Huiping Li, Weijun Feng, Xiu Xu","doi":"10.1016/j.ymthe.2024.08.017","DOIUrl":"10.1016/j.ymthe.2024.08.017","url":null,"abstract":"<p><p>The microcephaly-capillary malformation (MIC-CAP) syndrome is a life-threatening disease caused by biallelic mutations of the STAMBP gene, which encodes an endosomal deubiquitinating enzyme. To establish a suitable preclinical animal model for clinical therapeutic practice, we generated a central nervous system (CNS)-specific Stambp knockout mouse model (Stambp <sup>Sox1-cKO</sup>) that phenocopies Stambp null mice including progressive microcephaly, postnatal growth retardation and complete penetrance of preweaning death. In this MIC-CAP syndrome mouse model, early-onset neuronal death occurs specifically in the hippocampus and cortex, accompanied by aggregation of ubiquitinated proteins, and massive neuroinflammation. Importantly, neonatal AAV9-mediated gene supplementation of Stambp in the brain could significantly improve neurological defects, sustain growth, and prolong the lifespan of Stambp<sup>Sox1-cKO</sup> mice. Together, our findings reveal a central role of brain defects in the pathogenesis of STAMBP deficiency and provide preclinical evidence that postnatal gene replacement is an effective approach to cure the disease.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142018089","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
TALEN-edited allogeneic inducible dual CAR T cells enable effective targeting of solid tumors while mitigating off-tumor toxicity. TALEN编辑的异体诱导型双CAR T细胞可有效靶向实体瘤,同时减轻瘤外毒性。
IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-08-21 DOI: 10.1016/j.ymthe.2024.08.018
Sonal Dharani, Hana Cho, Jorge Postigo Fernandez, Alexandre Juillerat, Julien Valton, Philippe Duchateau, Laurent Poirot, Shipra Das

Adoptive cell therapy using chimeric antigen receptor (CAR) T cells has proven to be lifesaving for many cancer patients. However, its therapeutic efficacy has been limited in solid tumors. One key factor for this is cancer-associated fibroblasts (CAFs) that modulate the tumor microenvironment (TME) to inhibit T cell infiltration and induce "T cell dysfunction." Additionally, the sparsity of tumor-specific antigens (TSA) and expression of CAR-directed tumor-associated antigens (TAA) on normal tissues often results in "on-target off-tumor" cytotoxicity, raising safety concerns. Using TALEN-mediated gene editing, we present here an innovative CAR T cell engineering strategy to overcome these challenges. Our allogeneic "Smart CAR T cells" are designed to express a constitutive CAR, targeting FAP+ CAFs in solid tumors. Additionally, a second CAR targeting a TAA such as mesothelin is specifically integrated at a TCR signaling-inducible locus like PDCD1. FAPCAR-mediated CAF targeting induces expression of the mesothelin CAR, establishing an IF/THEN-gated circuit sensitive to dual antigen sensing. Using this approach, we observe enhanced anti-tumor cytotoxicity, while limiting "on-target off-tumor" toxicity. Our study thus demonstrates TALEN-mediated gene editing capabilities for design of allogeneic IF/THEN-gated dual CAR T cells that efficiently target immunotherapy-recalcitrant solid tumors while mitigating potential safety risks, encouraging clinical development of this strategy.

事实证明,使用嵌合抗原受体(CAR)T 细胞的适应性细胞疗法可以挽救许多癌症患者的生命。然而,它对实体瘤的疗效有限。其中一个关键因素是癌症相关成纤维细胞(CAFs),它们会调节肿瘤微环境(TME),抑制 T 细胞浸润并诱发 "T 细胞功能障碍"。此外,肿瘤特异性抗原(TSA)的稀缺性和CAR定向肿瘤相关抗原(TAA)在正常组织上的表达往往会导致 "靶外 "细胞毒性,从而引发安全性问题。利用 TALEN 介导的基因编辑,我们在此介绍一种创新的 CAR-T 细胞工程策略,以克服这些挑战。我们的异体 "智能 CAR T 细胞 "被设计为表达组成型 CAR,靶向实体瘤中的 FAP+ CAFs。此外,第二种靶向肿瘤相关抗原(TAA)(如间皮素)的CAR特异性地整合在TCR信号诱导位点(如PDCD1)上。FAPCAR 介导的 CAF 靶向会诱导间皮素 CAR 的表达,从而建立一个对双重抗原感应敏感的 IF/THEN 门控回路。利用这种方法,我们观察到了增强的抗肿瘤细胞毒性,同时限制了 "靶标外 "毒性。因此,我们的研究展示了 TALEN 介导的基因编辑能力,可用于设计异体 IF/THEN 门控双 CAR T 细胞,有效靶向免疫疗法再现性实体瘤,同时降低潜在的安全风险,从而鼓励这一策略的临床开发。
{"title":"TALEN-edited allogeneic inducible dual CAR T cells enable effective targeting of solid tumors while mitigating off-tumor toxicity.","authors":"Sonal Dharani, Hana Cho, Jorge Postigo Fernandez, Alexandre Juillerat, Julien Valton, Philippe Duchateau, Laurent Poirot, Shipra Das","doi":"10.1016/j.ymthe.2024.08.018","DOIUrl":"10.1016/j.ymthe.2024.08.018","url":null,"abstract":"<p><p>Adoptive cell therapy using chimeric antigen receptor (CAR) T cells has proven to be lifesaving for many cancer patients. However, its therapeutic efficacy has been limited in solid tumors. One key factor for this is cancer-associated fibroblasts (CAFs) that modulate the tumor microenvironment (TME) to inhibit T cell infiltration and induce \"T cell dysfunction.\" Additionally, the sparsity of tumor-specific antigens (TSA) and expression of CAR-directed tumor-associated antigens (TAA) on normal tissues often results in \"on-target off-tumor\" cytotoxicity, raising safety concerns. Using TALEN-mediated gene editing, we present here an innovative CAR T cell engineering strategy to overcome these challenges. Our allogeneic \"Smart CAR T cells\" are designed to express a constitutive CAR, targeting FAP<sup>+</sup> CAFs in solid tumors. Additionally, a second CAR targeting a TAA such as mesothelin is specifically integrated at a TCR signaling-inducible locus like PDCD1. FAPCAR-mediated CAF targeting induces expression of the mesothelin CAR, establishing an IF/THEN-gated circuit sensitive to dual antigen sensing. Using this approach, we observe enhanced anti-tumor cytotoxicity, while limiting \"on-target off-tumor\" toxicity. Our study thus demonstrates TALEN-mediated gene editing capabilities for design of allogeneic IF/THEN-gated dual CAR T cells that efficiently target immunotherapy-recalcitrant solid tumors while mitigating potential safety risks, encouraging clinical development of this strategy.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142018092","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effective knockdown-replace gene therapy in a novel mouse model of DNM1 developmental and epileptic encephalopathy. 一种新型 DNM1 发育和癫痫脑病小鼠模型的有效基因敲除-替代基因疗法。
IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-08-10 DOI: 10.1016/j.ymthe.2024.08.009
Devin J Jones, Divya Soundararajan, Noah K Taylor, Osasumwen V Aimiuwu, Pranav Mathkar, Amy Shore, Jia Jie Teoh, Wanqi Wang, Tristan T Sands, Matthew C Weston, Scott Q Harper, Wayne N Frankel

Effective gene therapy for gain-of-function or dominant-negative disease mutations may require eliminating expression of the mutant copy together with wild-type replacement. We evaluated such a knockdown-replace strategy in a mouse model of DNM1 disease, a debilitating and intractable neurodevelopmental epilepsy. To challenge the approach robustly, we expressed a patient-based variant in GABAergic neurons-which resulted in growth delay and lethal seizures evident by postnatal week three-and delivered to newborn pups an AAV9-based vector encoding a ubiquitously expressed, Dnm1-specific interfering RNA (RNAi) bivalently in tail-to-tail configuration with a neuron-specific, RNAi-resistant, codon-optimized Dnm1 cDNA. Pups receiving RNAi or cDNA alone fared no better than untreated pups, whereas the vast majority of mutants receiving modest doses survived with almost full growth recovery. Synaptic recordings of cortical neurons derived from treated pups revealed that significant alterations in transmission from inhibitory to excitatory neurons were rectified by bivalent vector application. To examine the mutant transcriptome and impact of treatment, we used RNA sequencing and functional annotation clustering. Mutants displayed abnormal expression of more than 1,000 genes in highly significant and relevant functional clusters, clusters that were abrogated by treatment. Together these results suggest knockdown-replace as a potentially effective strategy for treating DNM1 and related genetic neurodevelopmental disease.

对功能增益或显性阴性疾病突变的有效基因治疗可能需要消除突变拷贝的表达,同时进行野生型替换。我们在 DNM1 疾病(一种使人衰弱的难治性神经发育性癫痫)小鼠模型中评估了这种 "敲除-替换 "策略。为了有力地挑战这种方法,我们在GABA能神经元中表达了一种基于患者的变体--这种变体会导致生长发育迟缓,并在出生后第三周出现致命性癫痫发作--并向新生幼鼠释放了一种基于AAV9的载体,这种载体编码了一种普遍表达的、Dnm1特异性干扰RNA(RNAi),这种RNAi与神经元特异性、抗RNAi、密码子优化的Dnm1 cDNA呈尾对尾双向配置。单独接受 RNAi 或 cDNA 的幼鼠的情况并不比未接受治疗的幼鼠好,而绝大多数接受适量剂量的突变体都存活了下来,几乎完全恢复了生长。对接受治疗的幼鼠皮质神经元进行的突触记录显示,抑制性神经元向兴奋性神经元的传导发生了显著变化,而二价载体的应用则纠正了这种变化。为了研究突变体转录组和治疗的影响,我们使用了 RNAseq 和功能注释聚类。突变体在高度重要和相关的功能集群中显示了超过 1000 个基因的异常表达,这些集群在治疗后消失。这些结果表明,基因敲除-置换是治疗 DNM1 和相关遗传性神经发育疾病的潜在有效策略。
{"title":"Effective knockdown-replace gene therapy in a novel mouse model of DNM1 developmental and epileptic encephalopathy.","authors":"Devin J Jones, Divya Soundararajan, Noah K Taylor, Osasumwen V Aimiuwu, Pranav Mathkar, Amy Shore, Jia Jie Teoh, Wanqi Wang, Tristan T Sands, Matthew C Weston, Scott Q Harper, Wayne N Frankel","doi":"10.1016/j.ymthe.2024.08.009","DOIUrl":"10.1016/j.ymthe.2024.08.009","url":null,"abstract":"<p><p>Effective gene therapy for gain-of-function or dominant-negative disease mutations may require eliminating expression of the mutant copy together with wild-type replacement. We evaluated such a knockdown-replace strategy in a mouse model of DNM1 disease, a debilitating and intractable neurodevelopmental epilepsy. To challenge the approach robustly, we expressed a patient-based variant in GABAergic neurons-which resulted in growth delay and lethal seizures evident by postnatal week three-and delivered to newborn pups an AAV9-based vector encoding a ubiquitously expressed, Dnm1-specific interfering RNA (RNAi) bivalently in tail-to-tail configuration with a neuron-specific, RNAi-resistant, codon-optimized Dnm1 cDNA. Pups receiving RNAi or cDNA alone fared no better than untreated pups, whereas the vast majority of mutants receiving modest doses survived with almost full growth recovery. Synaptic recordings of cortical neurons derived from treated pups revealed that significant alterations in transmission from inhibitory to excitatory neurons were rectified by bivalent vector application. To examine the mutant transcriptome and impact of treatment, we used RNA sequencing and functional annotation clustering. Mutants displayed abnormal expression of more than 1,000 genes in highly significant and relevant functional clusters, clusters that were abrogated by treatment. Together these results suggest knockdown-replace as a potentially effective strategy for treating DNM1 and related genetic neurodevelopmental disease.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1,"publicationDate":"2024-08-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141913352","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Small-molecule-based targeted therapy in liver cancer. 肝癌的小分子靶向治疗。
IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-08-08 DOI: 10.1016/j.ymthe.2024.08.001
Yue Ming, Yanqiu Gong, Xuewen Fu, Xinyu Ouyang, Yong Peng, Wenchen Pu

Liver cancer is one of the most prevalent malignant tumors worldwide. According to the Barcelona Clinic Liver Cancer staging criteria, clinical guidelines provide tutorials to clinical management of liver cancer at their individual stages. However, most patients diagnosed with liver cancer are at advanced stage; therefore, many researchers conduct investigations on targeted therapy, aiming to improve the overall survival of these patients. To date, small-molecule-based targeted therapies are highly recommended (first line: sorafenib and lenvatinib; second line: regorafenib and cabozantinib) by current the clinical guidelines of the American Society of Clinical Oncology, European Society for Medical Oncology, and National Comprehensive Cancer Network. Herein, we summarize the small-molecule-based targeted therapies in liver cancer, including the approved and preclinical therapies as well as the therapies under clinical trials, and introduce their history of discovery, clinical trials, indications, and molecular mechanisms. For drug resistance, the revealed mechanisms of action and the combination therapies are also discussed. In fact, the known small-molecule-based therapies still have limited clinical benefits to liver cancer patients. Therefore, we analyze the current status and give our ideas for the urgent issues and future directions in this field, suggesting clues for novel techniques in liver cancer treatment.

肝癌是全球发病率最高的恶性肿瘤之一。根据巴塞罗那临床肝癌分期标准,临床指南为肝癌各期的临床治疗提供了指导。然而,大多数肝癌患者确诊时已是晚期,因此,许多研究人员开展了靶向治疗研究,旨在提高这些患者的总体生存率。迄今为止,以小分子为基础的靶向治疗备受推崇(拳头产品:索拉非尼(Sorafenib)和伦伐替尼(Lenvatinib);二线药物目前,美国临床肿瘤学会、欧洲肿瘤内科学会和美国国家综合癌症网络的临床指南都强烈推荐小分子靶向疗法(一线:索拉非尼和伦伐替尼;二线:瑞戈非尼和卡博赞替尼)。在此,我们总结了肝癌的小分子靶向疗法,包括已批准的和临床前的疗法以及正在进行临床试验的疗法,并介绍了它们的发现历史、临床试验、适应症和分子机制。针对耐药性,还讨论了揭示的作用机制和联合疗法。事实上,已知的小分子疗法对肝癌患者的临床疗效仍然有限。因此,我们分析了该领域的现状,并提出了我们对该领域亟待解决的问题和未来发展方向的看法,为肝癌治疗的新技术提供了线索。
{"title":"Small-molecule-based targeted therapy in liver cancer.","authors":"Yue Ming, Yanqiu Gong, Xuewen Fu, Xinyu Ouyang, Yong Peng, Wenchen Pu","doi":"10.1016/j.ymthe.2024.08.001","DOIUrl":"10.1016/j.ymthe.2024.08.001","url":null,"abstract":"<p><p>Liver cancer is one of the most prevalent malignant tumors worldwide. According to the Barcelona Clinic Liver Cancer staging criteria, clinical guidelines provide tutorials to clinical management of liver cancer at their individual stages. However, most patients diagnosed with liver cancer are at advanced stage; therefore, many researchers conduct investigations on targeted therapy, aiming to improve the overall survival of these patients. To date, small-molecule-based targeted therapies are highly recommended (first line: sorafenib and lenvatinib; second line: regorafenib and cabozantinib) by current the clinical guidelines of the American Society of Clinical Oncology, European Society for Medical Oncology, and National Comprehensive Cancer Network. Herein, we summarize the small-molecule-based targeted therapies in liver cancer, including the approved and preclinical therapies as well as the therapies under clinical trials, and introduce their history of discovery, clinical trials, indications, and molecular mechanisms. For drug resistance, the revealed mechanisms of action and the combination therapies are also discussed. In fact, the known small-molecule-based therapies still have limited clinical benefits to liver cancer patients. Therefore, we analyze the current status and give our ideas for the urgent issues and future directions in this field, suggesting clues for novel techniques in liver cancer treatment.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141902422","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
T-cell specific in vivo gene delivery with DART-AAVs targeted to CD8. 利用针对 CD8 的 DART-AAVs 在体内进行特异性 T 细胞基因递送。
IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-08-08 DOI: 10.1016/j.ymthe.2024.08.002
Muhammed Burak Demircan, Luca J Zinser, Alexander Michels, Mar Guaza-Lasheras, Fabian John, Johanna M Gorol, Samuel A Theuerkauf, Dorothee M Günther, Dirk Grimm, Florian R Greten, Petr Chlanda, Frederic B Thalheimer, Christian J Buchholz

One of the biggest challenges for in vivo gene therapy are vectors mediating highly selective gene transfer into a defined population of therapy-relevant cells. Here we present DARPin-targeted AAVs (DART-AAVs) displaying DARPins specific for human and murine CD8. Insertion of DARPins into the GH2/GH3 loop of the capsid protein 1 (VP1) of AAV2 and AAV6 resulted in high selectivity for CD8-positive T cells with unimpaired gene delivery activity. Remarkably, the capsid core structure was unaltered with protruding DARPins detectable. In complex primary cell mixtures, including donor blood or systemic injections into mice, the CD8-targeted AAVs were by far superior to unmodified AAV2 and AAV6 in terms of selectivity, target cell viability, and gene transfer rates. In vivo, up to 80% of activated CD8+ T cells were hit upon a single vector injection into conditioned humanized or immunocompetent mice. While gene transfer rates decreased significantly under non-activated conditions, genomic modification selectively in CD8+ T cells was still detectable upon Cre delivery into indicator mice. In both mouse models, selectivity for CD8+ T cells was close to absolute with exceptional detargeting from liver. The CD8-AAVs described here expand strategies for immunological research and in vivo gene therapy options.

体内基因治疗面临的最大挑战之一是如何将高选择性基因转移到确定的治疗相关细胞群中。在这里,我们展示了DARPin靶向AAVs(DART-AAVs),它显示了特异于人类和小鼠CD8的DARPin。将 DARPins 插入 AAV2 和 AAV6 的囊膜蛋白 1 (VP1) 的 GH2/GH3 环后,CD8 阳性 T 细胞具有高度选择性,基因递送活性不受影响。值得注意的是,AAV2 和 AAV6 的噬菌体核心结构没有改变,而且还能检测到突出的 DARPins。在复杂的原代细胞混合物(包括供体血液或小鼠全身注射)中,CD8 靶向 AAV 在选择性、靶细胞活力和基因转移率方面远远优于未修饰的 AAV2 和 AAV6。在体内,向条件化人源化小鼠或免疫功能健全的小鼠注射一次载体后,高达 80% 的活化 CD8+ T 细胞被击中。虽然基因转移率在非活化条件下明显下降,但将 Cre 运送到指示性小鼠体内后,仍可检测到 CD8+ T 细胞中选择性的基因组修饰。在这两种小鼠模型中,CD8+ T 细胞的选择性接近绝对值,而且能从肝脏中脱靶。本文描述的 CD8-AAVs 扩展了免疫学研究和体内基因治疗的策略。
{"title":"T-cell specific in vivo gene delivery with DART-AAVs targeted to CD8.","authors":"Muhammed Burak Demircan, Luca J Zinser, Alexander Michels, Mar Guaza-Lasheras, Fabian John, Johanna M Gorol, Samuel A Theuerkauf, Dorothee M Günther, Dirk Grimm, Florian R Greten, Petr Chlanda, Frederic B Thalheimer, Christian J Buchholz","doi":"10.1016/j.ymthe.2024.08.002","DOIUrl":"10.1016/j.ymthe.2024.08.002","url":null,"abstract":"<p><p>One of the biggest challenges for in vivo gene therapy are vectors mediating highly selective gene transfer into a defined population of therapy-relevant cells. Here we present DARPin-targeted AAVs (DART-AAVs) displaying DARPins specific for human and murine CD8. Insertion of DARPins into the GH2/GH3 loop of the capsid protein 1 (VP1) of AAV2 and AAV6 resulted in high selectivity for CD8-positive T cells with unimpaired gene delivery activity. Remarkably, the capsid core structure was unaltered with protruding DARPins detectable. In complex primary cell mixtures, including donor blood or systemic injections into mice, the CD8-targeted AAVs were by far superior to unmodified AAV2 and AAV6 in terms of selectivity, target cell viability, and gene transfer rates. In vivo, up to 80% of activated CD8+ T cells were hit upon a single vector injection into conditioned humanized or immunocompetent mice. While gene transfer rates decreased significantly under non-activated conditions, genomic modification selectively in CD8+ T cells was still detectable upon Cre delivery into indicator mice. In both mouse models, selectivity for CD8+ T cells was close to absolute with exceptional detargeting from liver. The CD8-AAVs described here expand strategies for immunological research and in vivo gene therapy options.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141902423","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PDIA3 orchestrates effector T cell program by serving as a chaperone to facilitate the non-canonical nuclear import of STAT1 and PKM2. PDIA3 是一种伴侣蛋白,可促进 STAT1 和 PKM2 的非经典核导入,从而协调效应 T 细胞程序。
IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-08-07 Epub Date: 2024-05-31 DOI: 10.1016/j.ymthe.2024.05.038
Chun-Liang Yang, Fa-Xi Wang, Jia-Hui Luo, Shan-Jie Rong, Wan-Ying Lu, Qi-Jie Chen, Jun Xiao, Ting Wang, Dan-Ni Song, Jing Liu, Qian Mo, Shuo Li, Yu Chen, Ya-Nan Wang, Yan-Jun Liu, Tong Yan, Wei-Kuan Gu, Shu Zhang, Fei Xiong, Qi-Lin Yu, Zi-Yun Zhang, Ping Yang, Shi-Wei Liu, Decio Eizirik, Ling-Li Dong, Fei Sun, Cong-Yi Wang

Dysregulated T cell activation underpins the immunopathology of rheumatoid arthritis (RA), yet the machineries that orchestrate T cell effector program remain incompletely understood. Herein, we leveraged bulk and single-cell RNA sequencing data from RA patients and validated protein disulfide isomerase family A member 3 (PDIA3) as a potential therapeutic target. PDIA3 is remarkably upregulated in pathogenic CD4 T cells derived from RA patients and positively correlates with C-reactive protein level and disease activity score 28. Pharmacological inhibition or genetic ablation of PDIA3 alleviates RA-associated articular pathology and autoimmune responses. Mechanistically, T cell receptor signaling triggers intracellular calcium flux to activate NFAT1, a process that is further potentiated by Wnt5a under RA settings. Activated NFAT1 then directly binds to the Pdia3 promoter to enhance the expression of PDIA3, which complexes with STAT1 or PKM2 to facilitate their nuclear import for transcribing T helper 1 (Th1) and Th17 lineage-related genes, respectively. This non-canonical regulatory mechanism likely occurs under pathological conditions, as PDIA3 could only be highly induced following aberrant external stimuli. Together, our data support that targeting PDIA3 is a vital strategy to mitigate autoimmune diseases, such as RA, in clinical settings.

T细胞活化失调是类风湿性关节炎(RA)免疫病理的基础,但人们对T细胞效应程序的协调机制仍不甚了解。在这里,我们利用来自类风湿性关节炎患者的大量和单细胞RNA测序数据,验证了蛋白二硫化物异构酶A3(PDIA3)是一个潜在的治疗靶点。PDIA3在来自RA患者的致病性CD4 T细胞中显著上调,并与C反应蛋白(CRP)水平和疾病活动评分28(DAS28)呈正相关。药物抑制或基因消融 PDIA3 可减轻 RA 相关的关节病变和自身免疫反应。从机理上讲,T细胞受体(TCR)信号触发细胞内钙通量以激活NFAT1,在RA环境下,Wnt5a进一步增强了这一过程。活化的 NFAT1 然后直接与 Pdia3 启动子结合,增强 PDIA3 的表达,PDIA3 与 STAT1 或 PKM2 复合物,促进它们的核输入,分别转录 Th1 和 Th17 系相关基因。这种非经典调控机制很可能发生在病理条件下,因为只有在异常外部刺激下,PDIA3 才会被高度诱导。总之,我们的数据支持靶向 PDIA3 是在临床环境中缓解自身免疫性疾病(如 RA)的重要策略。
{"title":"PDIA3 orchestrates effector T cell program by serving as a chaperone to facilitate the non-canonical nuclear import of STAT1 and PKM2.","authors":"Chun-Liang Yang, Fa-Xi Wang, Jia-Hui Luo, Shan-Jie Rong, Wan-Ying Lu, Qi-Jie Chen, Jun Xiao, Ting Wang, Dan-Ni Song, Jing Liu, Qian Mo, Shuo Li, Yu Chen, Ya-Nan Wang, Yan-Jun Liu, Tong Yan, Wei-Kuan Gu, Shu Zhang, Fei Xiong, Qi-Lin Yu, Zi-Yun Zhang, Ping Yang, Shi-Wei Liu, Decio Eizirik, Ling-Li Dong, Fei Sun, Cong-Yi Wang","doi":"10.1016/j.ymthe.2024.05.038","DOIUrl":"10.1016/j.ymthe.2024.05.038","url":null,"abstract":"<p><p>Dysregulated T cell activation underpins the immunopathology of rheumatoid arthritis (RA), yet the machineries that orchestrate T cell effector program remain incompletely understood. Herein, we leveraged bulk and single-cell RNA sequencing data from RA patients and validated protein disulfide isomerase family A member 3 (PDIA3) as a potential therapeutic target. PDIA3 is remarkably upregulated in pathogenic CD4 T cells derived from RA patients and positively correlates with C-reactive protein level and disease activity score 28. Pharmacological inhibition or genetic ablation of PDIA3 alleviates RA-associated articular pathology and autoimmune responses. Mechanistically, T cell receptor signaling triggers intracellular calcium flux to activate NFAT1, a process that is further potentiated by Wnt5a under RA settings. Activated NFAT1 then directly binds to the Pdia3 promoter to enhance the expression of PDIA3, which complexes with STAT1 or PKM2 to facilitate their nuclear import for transcribing T helper 1 (Th1) and Th17 lineage-related genes, respectively. This non-canonical regulatory mechanism likely occurs under pathological conditions, as PDIA3 could only be highly induced following aberrant external stimuli. Together, our data support that targeting PDIA3 is a vital strategy to mitigate autoimmune diseases, such as RA, in clinical settings.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1,"publicationDate":"2024-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141186813","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Building a novel TRUCK by harnessing the endogenous IFN-gamma promoter for cytokine expression. 利用内源性 IFN-gamma 启动子表达细胞因子,构建新型 TRUCK。
IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-08-07 Epub Date: 2024-06-15 DOI: 10.1016/j.ymthe.2024.06.017
Liya Ma, Kaiwen Zhang, Jian Xu, Jian Wang, Ting Jiang, Xiaolong Du, Jiaxin Zhang, Jing Huang, Fengyi Ren, Dong Liu, Weiwei Xue, Dongxu Kan, Mengjiao Yao, Yutian Liang, Hongxing Jason-Sun

Despite the remarkable success of chimeric antigen receptor (CAR) T therapy in hematological malignancies, its efficacy in solid tumors remains limited. Cytokine-engineered CAR T cells offer a promising avenue, yet their clinical translation is hindered by the risks associated with constitutive cytokine expression. In this proof-of-concept study, we leverage the endogenous interferon (IFN)-γ promoter for transgenic interleukin (IL)-15 expression. We demonstrate that IFN-γ expression is tightly regulated by T cell receptor signaling. By introducing an internal ribosome entry site IL15 into the 3' UTR of the IFN-γ gene via homology directed repair-mediated knock-in, we confirm that IL-15 expression can co-express with IFN-γ in an antigen stimulation-dependent manner. Importantly, the insertion of transgenes does not compromise endogenous IFN-γ expression. In vitro and in vivo data demonstrate that IL-15 driven by the IFN-γ promoter dramatically improves CAR T cells' antitumor activity, suggesting the effectiveness of IL-15 expression. Last, as a part of our efforts toward clinical translation, we have developed an innovative two-gene knock-in approach. This approach enables the simultaneous integration of CAR and IL-15 genes into TRAC and IFN-γ gene loci using a single AAV vector. CAR T cells engineered to express IL-15 using this approach demonstrate enhanced antitumor efficacy. Overall, our study underscores the feasibility of utilizing endogenous promoters for transgenic cytokines expression in CAR T cells.

尽管 CAR T 疗法在血液恶性肿瘤中取得了巨大成功,但其在实体瘤中的疗效仍然有限。细胞因子工程化的 CAR T 细胞提供了一条前景广阔的途径,但其临床转化却受到组成型细胞因子表达相关风险的阻碍。在这项概念验证研究中,我们利用内源性 IFN-γ 启动子进行转基因 IL-15 表达。我们证明 IFN-γ 的表达受到 TCR 信号的严格调控。通过 HDR 介导的基因敲入将 IRES-IL15 导入 IFN-γ 基因的 3'-UTR 中,我们证实 IL-15 的表达可与 IFN-γ 以抗原刺激依赖性的方式共同表达。重要的是,转基因的插入不会影响内源性 IFN-γ 的表达。体外和体内数据表明,IFN-γ 启动子驱动的 IL-15 能显著提高 CAR T 细胞的抗肿瘤活性,这表明 IL-15 的表达是有效的。最后,作为临床转化努力的一部分,我们开发了一种创新的双基因敲入方法。这种方法使用单一 AAV 载体将 CAR 和 IL-15 基因同时整合到 TRAC 和 IFN-γ 基因座中。使用这种方法设计的表达 IL-15 的 CAR T 细胞显示出更强的抗肿瘤功效。总之,我们的研究强调了利用内源性启动子在 CAR T 细胞中表达转基因细胞因子的可行性。
{"title":"Building a novel TRUCK by harnessing the endogenous IFN-gamma promoter for cytokine expression.","authors":"Liya Ma, Kaiwen Zhang, Jian Xu, Jian Wang, Ting Jiang, Xiaolong Du, Jiaxin Zhang, Jing Huang, Fengyi Ren, Dong Liu, Weiwei Xue, Dongxu Kan, Mengjiao Yao, Yutian Liang, Hongxing Jason-Sun","doi":"10.1016/j.ymthe.2024.06.017","DOIUrl":"10.1016/j.ymthe.2024.06.017","url":null,"abstract":"<p><p>Despite the remarkable success of chimeric antigen receptor (CAR) T therapy in hematological malignancies, its efficacy in solid tumors remains limited. Cytokine-engineered CAR T cells offer a promising avenue, yet their clinical translation is hindered by the risks associated with constitutive cytokine expression. In this proof-of-concept study, we leverage the endogenous interferon (IFN)-γ promoter for transgenic interleukin (IL)-15 expression. We demonstrate that IFN-γ expression is tightly regulated by T cell receptor signaling. By introducing an internal ribosome entry site IL15 into the 3' UTR of the IFN-γ gene via homology directed repair-mediated knock-in, we confirm that IL-15 expression can co-express with IFN-γ in an antigen stimulation-dependent manner. Importantly, the insertion of transgenes does not compromise endogenous IFN-γ expression. In vitro and in vivo data demonstrate that IL-15 driven by the IFN-γ promoter dramatically improves CAR T cells' antitumor activity, suggesting the effectiveness of IL-15 expression. Last, as a part of our efforts toward clinical translation, we have developed an innovative two-gene knock-in approach. This approach enables the simultaneous integration of CAR and IL-15 genes into TRAC and IFN-γ gene loci using a single AAV vector. CAR T cells engineered to express IL-15 using this approach demonstrate enhanced antitumor efficacy. Overall, our study underscores the feasibility of utilizing endogenous promoters for transgenic cytokines expression in CAR T cells.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1,"publicationDate":"2024-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141327697","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Clinical perspectives: Treating spinal muscular atrophy. 临床视角:治疗脊髓肌肉萎缩症。
IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-08-07 Epub Date: 2024-06-18 DOI: 10.1016/j.ymthe.2024.06.020
Molly A McPheron, Marcia V Felker

Spinal muscular atrophy is a rare and progressive neuromuscular disease that, without treatment, leads to progressive weakness and often death. A plethora of studies have led to the approval of three high-cost and effective treatments since 2016. These treatments, nusinersen, onasemnogene abeparvovec, and risdiplam, have not been directly compared and have varying challenges in administration. In this review, we discuss the evidence supporting the use of these medications, the process of treatment selection, monitoring after treatment, the limited data comparing treatments, as well as future directions for investigation and therapy.

脊髓性肌萎缩症是一种罕见的渐进性神经肌肉疾病,如不及时治疗,会导致患者逐渐虚弱,甚至死亡。自2016年以来,大量研究促使三种高成本、有效的治疗方法获得批准。这些疗法,即努西奈森、onasemnogene abeparvovec和利迪普兰,尚未进行过直接比较,在用药方面也存在不同的挑战。在这篇综述中,我们将讨论支持使用这些药物的证据、治疗选择过程、治疗后的监测、比较治疗的有限数据以及未来的研究和治疗方向。
{"title":"Clinical perspectives: Treating spinal muscular atrophy.","authors":"Molly A McPheron, Marcia V Felker","doi":"10.1016/j.ymthe.2024.06.020","DOIUrl":"10.1016/j.ymthe.2024.06.020","url":null,"abstract":"<p><p>Spinal muscular atrophy is a rare and progressive neuromuscular disease that, without treatment, leads to progressive weakness and often death. A plethora of studies have led to the approval of three high-cost and effective treatments since 2016. These treatments, nusinersen, onasemnogene abeparvovec, and risdiplam, have not been directly compared and have varying challenges in administration. In this review, we discuss the evidence supporting the use of these medications, the process of treatment selection, monitoring after treatment, the limited data comparing treatments, as well as future directions for investigation and therapy.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1,"publicationDate":"2024-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141419957","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
An aptamer-mediated base editing platform for simultaneous knockin and multiple gene knockout for allogeneic CAR-T cells generation. 一种以适配体为介导的碱基编辑平台,用于同时敲入和敲除多个基因,以生成异体 CAR-T 细胞。
IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-08-07 Epub Date: 2024-06-26 DOI: 10.1016/j.ymthe.2024.06.033
Immacolata Porreca, Robert Blassberg, Jennifer Harbottle, Bronwyn Joubert, Olga Mielczarek, Jesse Stombaugh, Kevin Hemphill, Jonathan Sumner, Deividas Pazeraitis, Julia Liz Touza, Margherita Francescatto, Mike Firth, Tommaso Selmi, Juan Carlos Collantes, Zaklina Strezoska, Benjamin Taylor, Shengkan Jin, Ceri M Wiggins, Anja van Brabant Smith, John J Lambourne

Gene editing technologies hold promise for enabling the next generation of adoptive cellular therapies. In conventional gene editing platforms that rely on nuclease activity, such as clustered regularly interspaced short palindromic repeats CRISPR-associated protein 9 (CRISPR-Cas9), allow efficient introduction of genetic modifications; however, these modifications occur via the generation of DNA double-strand breaks (DSBs) and can lead to unwanted genomic alterations and genotoxicity. Here, we apply a novel modular RNA aptamer-mediated Pin-point base editing platform to simultaneously introduce multiple gene knockouts and site-specific integration of a transgene in human primary T cells. We demonstrate high editing efficiency and purity at all target sites and significantly reduced frequency of chromosomal translocations compared with the conventional CRISPR-Cas9 system. Site-specific knockin of a chimeric antigen receptor and multiplex gene knockout are achieved within a single intervention and without the requirement for additional sequence-targeting components. The ability to perform complex genome editing efficiently and precisely highlights the potential of the Pin-point platform for application in a range of advanced cell therapies.

基因编辑技术有望实现下一代的采纳性细胞疗法。传统的基因编辑平台依赖于核酸酶活性,例如聚类规则间隔短回文重复序列-CRISPR相关蛋白9(CRISPR-Cas9),可以有效地引入基因修饰;然而,这些修饰是通过产生DNA双链断裂(DSB)发生的,可能会导致不必要的基因组改变和基因毒性。在这里,我们应用了一种新型模块化 RNA aptamer 介导的 Pin-point™ 碱基编辑平台,在人类原代 T 细胞中同时引入多个基因敲除和转基因的特异性位点整合。与传统的 CRISPR-Cas9 系统相比,我们在所有目标位点都证明了很高的编辑效率和纯度,并显著降低了染色体易位的频率。一次干预即可实现嵌合抗原受体(CAR)的特异位点敲入和多重基因敲除,无需额外的序列靶向元件。高效、精确地进行复杂基因组编辑的能力,凸显了 Pin-point 平台在一系列先进细胞疗法中的应用潜力。
{"title":"An aptamer-mediated base editing platform for simultaneous knockin and multiple gene knockout for allogeneic CAR-T cells generation.","authors":"Immacolata Porreca, Robert Blassberg, Jennifer Harbottle, Bronwyn Joubert, Olga Mielczarek, Jesse Stombaugh, Kevin Hemphill, Jonathan Sumner, Deividas Pazeraitis, Julia Liz Touza, Margherita Francescatto, Mike Firth, Tommaso Selmi, Juan Carlos Collantes, Zaklina Strezoska, Benjamin Taylor, Shengkan Jin, Ceri M Wiggins, Anja van Brabant Smith, John J Lambourne","doi":"10.1016/j.ymthe.2024.06.033","DOIUrl":"10.1016/j.ymthe.2024.06.033","url":null,"abstract":"<p><p>Gene editing technologies hold promise for enabling the next generation of adoptive cellular therapies. In conventional gene editing platforms that rely on nuclease activity, such as clustered regularly interspaced short palindromic repeats CRISPR-associated protein 9 (CRISPR-Cas9), allow efficient introduction of genetic modifications; however, these modifications occur via the generation of DNA double-strand breaks (DSBs) and can lead to unwanted genomic alterations and genotoxicity. Here, we apply a novel modular RNA aptamer-mediated Pin-point base editing platform to simultaneously introduce multiple gene knockouts and site-specific integration of a transgene in human primary T cells. We demonstrate high editing efficiency and purity at all target sites and significantly reduced frequency of chromosomal translocations compared with the conventional CRISPR-Cas9 system. Site-specific knockin of a chimeric antigen receptor and multiplex gene knockout are achieved within a single intervention and without the requirement for additional sequence-targeting components. The ability to perform complex genome editing efficiently and precisely highlights the potential of the Pin-point platform for application in a range of advanced cell therapies.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1,"publicationDate":"2024-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141469643","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Human plasma cells engineered to secrete bispecifics drive effective in vivo leukemia killing. 经改造可分泌双特异性药物的人浆细胞可在体内有效杀死白血病。
IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Pub Date : 2024-08-07 Epub Date: 2024-07-02 DOI: 10.1016/j.ymthe.2024.06.004
Tyler F Hill, Parnal Narvekar, Gregory D Asher, Jasmine N Edelstein, Nathan D Camp, Annaiz Grimm, Kerri R Thomas, Michael D Leiken, Katherine M Molloy, Peter J Cook, Sean P Arlauckas, Richard A Morgan, Sarah K Tasian, David J Rawlings, Richard G James

Bispecific antibodies are an important tool for the management and treatment of acute leukemias. As a next step toward clinical translation of engineered plasma cells, we describe approaches for secretion of bispecific antibodies by human plasma cells. We show that human plasma cells expressing either fragment crystallizable domain-deficient anti-CD19 × anti-CD3 (blinatumomab) or anti-CD33 × anti-CD3 bispecific antibodies mediate T cell activation and direct T cell killing of B acute lymphoblastic leukemia or acute myeloid leukemia cell lines in vitro. We demonstrate that knockout of the self-expressed antigen, CD19, boosts anti-CD19-bispecific secretion by plasma cells and prevents self-targeting. Plasma cells secreting anti-CD19-bispecific antibodies elicited in vivo control of acute lymphoblastic leukemia patient-derived xenografts in immunodeficient mice co-engrafted with autologous T cells. In these studies, we found that leukemic control elicited by engineered plasma cells was similar to CD19-targeted chimeric antigen receptor-expressing T cells. Finally, the steady-state concentration of anti-CD19 bispecifics in serum 1 month after cell delivery and tumor eradication was comparable with that observed in patients treated with a steady-state infusion of blinatumomab. These findings support further development of ePCs for use as a durable delivery system for the treatment of acute leukemias, and potentially other cancers.

双特异性抗体是管理和治疗急性白血病的重要工具。作为工程浆细胞临床转化的下一步,我们描述了人类浆细胞分泌双特异性抗体的方法。我们发现,表达片段可结晶结构域缺陷的抗 CD19 × 抗 CD3(blinatumomab)或抗 CD33 × 抗 CD3 双特异性抗体的人浆细胞能在体外介导 T 细胞活化并直接杀伤 B 型急性淋巴细胞白血病或急性髓性白血病细胞系。我们证明,敲除自身表达的抗原 CD19 会促进浆细胞分泌抗 CD19 双特异性抗体,并阻止自身靶向。分泌抗 CD19 双特异性抗体的浆细胞在免疫缺陷小鼠体内控制了与自体 T 细胞共同移植的急性淋巴细胞白血病患者衍生的异种移植物。在这些研究中,我们发现工程浆细胞诱导的白血病控制与 CD19 靶向嵌合抗原受体表达 T 细胞相似。最后,细胞递送和肿瘤根除1个月后,血清中抗CD19双特异性的稳态浓度与稳态输注blinatumomab治疗患者的浓度相当。这些研究结果支持进一步开发ePCs,将其用作治疗急性白血病和其他潜在癌症的持久递送系统。
{"title":"Human plasma cells engineered to secrete bispecifics drive effective in vivo leukemia killing.","authors":"Tyler F Hill, Parnal Narvekar, Gregory D Asher, Jasmine N Edelstein, Nathan D Camp, Annaiz Grimm, Kerri R Thomas, Michael D Leiken, Katherine M Molloy, Peter J Cook, Sean P Arlauckas, Richard A Morgan, Sarah K Tasian, David J Rawlings, Richard G James","doi":"10.1016/j.ymthe.2024.06.004","DOIUrl":"10.1016/j.ymthe.2024.06.004","url":null,"abstract":"<p><p>Bispecific antibodies are an important tool for the management and treatment of acute leukemias. As a next step toward clinical translation of engineered plasma cells, we describe approaches for secretion of bispecific antibodies by human plasma cells. We show that human plasma cells expressing either fragment crystallizable domain-deficient anti-CD19 × anti-CD3 (blinatumomab) or anti-CD33 × anti-CD3 bispecific antibodies mediate T cell activation and direct T cell killing of B acute lymphoblastic leukemia or acute myeloid leukemia cell lines in vitro. We demonstrate that knockout of the self-expressed antigen, CD19, boosts anti-CD19-bispecific secretion by plasma cells and prevents self-targeting. Plasma cells secreting anti-CD19-bispecific antibodies elicited in vivo control of acute lymphoblastic leukemia patient-derived xenografts in immunodeficient mice co-engrafted with autologous T cells. In these studies, we found that leukemic control elicited by engineered plasma cells was similar to CD19-targeted chimeric antigen receptor-expressing T cells. Finally, the steady-state concentration of anti-CD19 bispecifics in serum 1 month after cell delivery and tumor eradication was comparable with that observed in patients treated with a steady-state infusion of blinatumomab. These findings support further development of ePCs for use as a durable delivery system for the treatment of acute leukemias, and potentially other cancers.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1,"publicationDate":"2024-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141498521","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","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
Molecular Therapy
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