safety switch

安全开关
  • 文章类型: Journal Article
    激活条件安全开关有可能逆转由工程细胞疗法引起的严重毒性,包括嵌合抗原受体(CAR)T细胞。功能惰性的,来自人表皮生长因子受体(EGFRt)的非免疫原性细胞表面标志物是一种有前途的安全开关,已用于多种临床构建体,可被西妥昔单抗靶向,临床上可用的单克隆抗体。然而,这种方法需要EGFRt的高的和持久的细胞表面表达,以确保抗体介导的工程细胞的清除是快速和完整的.在这里,我们表明将短的近膜序列掺入EGFRt多肽可增强其在T细胞表面的表达及其对抗体依赖性细胞毒性(ADCC)的敏感性。与EGFRt相比,将该优化变体(EGFRopt)并入双顺反子和三顺反子CAR设计中导致CART细胞的更快速的体内消除和它们的效应子活性的稳健终止。这些研究将EGFRopt确立为下一代基于细胞的疗法开发的卓越安全开关。
    Activation of a conditional safety switch has the potential to reverse serious toxicities arising from the administration of engineered cellular therapies, including chimeric antigen receptor (CAR) T cells. The functionally inert, non-immunogenic cell surface marker derived from human epidermal growth factor receptor (EGFRt) is a promising safety switch that has been used in multiple clinical constructs and can be targeted by cetuximab, a clinically available monoclonal antibody. However, this approach requires high and persistent cell surface expression of EGFRt to ensure that antibody-mediated depletion of engineered cells is rapid and complete. Here we show that incorporating a short juxtamembrane sequence into the EGFRt polypeptide enhances its expression on the surface of T cells and their susceptibility to antibody-dependent cellular cytotoxicity (ADCC). Incorporating this optimized variant (EGFRopt) into bicistronic and tricistronic CAR designs results in more rapid in vivo elimination of CAR T cells and robust termination of their effector activity compared to EGFRt. These studies establish EGFRopt as a superior safety switch for the development of next-generation cell-based therapeutics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    干细胞和祖细胞为再生医学和基因治疗方法带来了巨大的希望。然而,活细胞的移植带来了有害生长的根本风险,CRISPR-Cas9或其他遗传操作可能会加剧。这里,我们描述了一种安全系统来控制细胞增殖,同时允许强大而有效的细胞制造,没有任何额外的遗传因素。停用TYMS,一种关键的核苷酸代谢酶,在几种细胞系中,导致细胞仅在补充外源性胸苷时才增殖。根据补充,TYMS-/-多能干细胞增殖,产生畸胎瘤并成功分化为潜在的治疗细胞类型,如胰腺β细胞。我们的结果表明,补充外源性胸苷会影响干细胞增殖,但不是干细胞衍生细胞的功能。分化后,有丝分裂后细胞在体外或体内不需要胸苷,如在植入干细胞来源的胰腺组织后长达5个月的小鼠中功能性人胰岛素的产生所示。
    Stem and progenitor cells hold great promise for regenerative medicine and gene therapy approaches. However, transplantation of living cells entails a fundamental risk of unwanted growth, potentially exacerbated by CRISPR-Cas9 or other genetic manipulations. Here, we describe a safety system to control cell proliferation while allowing robust and efficient cell manufacture, without any added genetic elements. Inactivating TYMS, a key nucleotide metabolism enzyme, in several cell lines resulted in cells that proliferate only when supplemented with exogenous thymidine. Under supplementation, TYMS-/--pluripotent stem cells proliferate, produce teratomas, and successfully differentiate into potentially therapeutic cell types such as pancreatic β cells. Our results suggest that supplementation with exogenous thymidine affects stem cell proliferation, but not the function of stem cell-derived cells. After differentiation, postmitotic cells do not require thymidine in vitro or in vivo, as shown by the production of functional human insulin in mice up to 5 months after implantation of stem cell-derived pancreatic tissue.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    嵌合抗原受体T细胞(CAR-T)疗法是一种创新的免疫治疗方法,其利用与T淋巴细胞的有效细胞毒性偶联的单克隆抗体(mAb)的特异性,利用遗传修饰的T细胞来消除癌细胞。CAR-T疗法在复发性/难治性B细胞恶性肿瘤中产生了显着改善。鉴于这些成功,CAR-T已迅速扩散到其他血液系统恶性肿瘤,并且在实体瘤中的研究越来越多。从早期临床应用到现在,CAR-T细胞疗法伴随着显著的毒性,即细胞因子释放综合征(CRS),免疫效应细胞相关神经毒性综合征(ICANS),和靶肿瘤外(OTOT)效应。虽然CRS和ICANS的医疗管理有所改善,难治性症状和未预期的特异性毒性的持续威胁凸显了需要更强有力的安全措施.随着CAR-T细胞疗法继续扩展到实体瘤领域,这尤其令人痛苦,不可预测的毒性风险仍然很高。我们将回顾CAR-T作为一种免疫治疗方法,包括在整个开发过程中出现独特的毒性。我们将讨论已知的和新的策略,以减轻这些毒性;在实体瘤的治疗额外的安全挑战,以及诱导型Caspase9“安全开关”如何为继续探索提供理想的平台。
    Chimeric antigen receptor T cell (CAR-T) therapy is an innovative immunotherapeutic approach that utilizes genetically modified T-cells to eliminate cancer cells using the specificity of a monoclonal antibody (mAb) coupled to the potent cytotoxicity of the T-lymphocyte. CAR-T therapy has yielded significant improvements in relapsed/refractory B-cell malignancies. Given these successes, CAR-T has quickly spread to other hematologic malignancies and is being increasingly explored in solid tumors. From early clinical applications to present day, CAR-T cell therapy has been accompanied by significant toxicities, namely cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), and on-target off-tumor (OTOT) effects. While medical management has improved for CRS and ICANS, the ongoing threat of refractory symptoms and unanticipated idiosyncratic toxicities highlights the need for more powerful safety measures. This is particularly poignant as CAR T-cell therapy continues to expand into the solid tumor space, where the risk of unpredictable toxicities remains high. We will review CAR-T as an immunotherapeutic approach including emergence of unique toxicities throughout development. We will discuss known and novel strategies to mitigate these toxicities; additional safety challenges in the treatment of solid tumors, and how the inducible Caspase 9 \"safety switch\" provides an ideal platform for continued exploration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    基因编辑和工程化的基于细胞的疗法是治疗各种疾病的有前途的方法,包括癌症.然而,工程化细胞能够长期持续存在的能力以及可能的毒性引起了人们对这些方法安全性的担忧.尽管许多不同的一维自杀系统已被纳入治疗细胞类型,需要结合允许在不同时间点控制杀伤治疗性细胞的双层自杀系统。在这项研究中,我们设计了多种治疗细胞来表达两种不同的杀伤开关,RapaCasp9和HSV-TK分别利用雷帕霉素和更昔洛韦激活这些杀伤开关。我们表明RapaCasp9和HSV-TK分子的功能被保留,并且可以被激活以分别在激活后早期(24小时)和晚期(48小时)检测到凋亡。没有毒性。在体内,我们显示了在皮下和原位模型中治疗后大部分细胞的根除。此外,我们演示了两个自杀开关如何独立工作,并且可以顺序激活以改善杀戮,从而确保在单个电池的激活不足以消除电池的情况下的故障保护机制。我们的发现强调了双重自杀系统的可靠性,对各种具有不同生物学特性的细胞有效,独立于他们的解剖存在。
    Gene edited and engineered cell-based therapies are a promising approach for treating a variety of disorders, including cancer. However, the ability of engineered cells to persist for prolonged periods along with possible toxicity raises concerns over the safety of these approaches. Although a number of different one-dimensional suicide systems have been incorporated into therapeutic cell types, the incorporation of a two-layered suicide system that allows controlled killing of therapeutic cells at different time points is needed. In this study, we engineered a variety of therapeutic cells to express two different kill switches, RapaCasp9 and HSV-TK and utilized Rapamycin and Ganciclovir respectively to activate these kill switches. We show that the function of both RapaCasp9 and HSV-TK molecules is preserved and can be activated to induce apoptosis detected early (24 h) and late (48 h) post-activation respectively, with no toxicity. In vivo, we show the eradication of a majority of cells after treatment in subcutaneous and orthotopic models. Furthermore, we demonstrate how both suicide switches work independently and can be activated sequentially for an improved killing, thus ensuring a failsafe mechanism in case the activation of a single one of them is not sufficient to eliminate the cells. Our findings highlight the reliability of the double suicide system, effective on a variety of cells with different biological characteristics, independent of their anatomic presence.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    基因修饰细胞疗法具有严重和潜在致命不良事件的固有风险。包括同种反应性细胞的扩增或由于插入诱变引起的恶性转化。减轻基因修饰细胞不受控制的增殖的策略包括共转染自杀基因,例如诱导型胱天蛋白酶9安全开关(ΔiC9)。然而,ΔiC9的激活不能完全消除所有基因修饰的细胞。因此,我们测试了一个独立或一起使用的两个自杀基因系统,以完全消除细胞为目标。第一种方法将ΔiC9与缺乏内源前结构域的诱导型半胱天冬酶8ΔiC8组合。理由是使用具有替代和互补作用机制的第二半胱天冬酶。Jurkat细胞共转导以共表达可由BB同源二聚体激活的ΔiC8,在模型中使用雷帕霉素类似物西罗莫司可激活的ΔiC9来估计诱导型细胞消除的程度。我们发现这两种药物都可以独立激活每种caspase,当两个系统同时激活时,基因修饰细胞的消除增强,细胞再生长明显减少。并行采用了第二种方法,将ΔiC9与RQR8紧凑型自杀基因相结合。RQR8包含CD20模拟表位,以抗CD20单克隆抗体利妥昔单抗为目标,和QBend10,ΔCD34选择标记。同样,当两个系统一起激活时,观察到细胞消除增强,细胞再生长明显减少。使用BB同二聚化剂也注意到剂量滴定效应,而西罗莫司在最低浓度下仍然非常有效。需要进一步的体内研究来验证这些新颖的组合系统,这可能在未来的癌症治疗或再生医学中发挥作用。
    Gene-modified cellular therapies carry inherent risks of severe and potentially fatal adverse events, including the expansion of alloreactive cells or malignant transformation due to insertional mutagenesis. Strategies to mitigate uncontrolled proliferation of gene-modified cells include co-transfection of a suicide gene, such as the inducible caspase 9 safety switch (ΔiC9). However, the activation of the ΔiC9 fails to completely eliminate all gene-modified cells. Therefore, we tested a two suicide gene system used independently or together, with the goal of complete cell elimination. The first approach combined the ΔiC9 with an inducible caspase 8, ΔiC8, which lacks the endogenous prodomain. The rationale was to use a second caspase with an alternative and complementary mechanism of action. Jurkat cells co-transduced to co-express the ΔiC8, activatable by a BB homodimerizer, and the ΔiC9 activatable by the rapamycin analog sirolimus were used in a model to estimate the degree of inducible cell elimination. We found that both agents could activate each caspase independently, with enhanced elimination with superior reduction in cell regrowth of gene-modified cells when both systems were activated simultaneously. A second approach was employed in parallel, combining the ΔiC9 with the RQR8 compact suicide gene. RQR8 incorporates a CD20 mimotope, targeted by the anti-CD20 monoclonal antibody rituxan, and the QBend10, a ΔCD34 selectable marker. Likewise, enhanced cell elimination with superior reduction in cell regrowth was observed when both systems were activated together. A dose-titration effect was also noted utilizing the BB homodimerizer, whereas sirolimus remained very potent at minimal concentrations. Further in vivo studies are needed to validate these novel combination systems, which may play a role in future cancer therapies or regenerative medicine.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    人类间充质干细胞(MSC)是多能干细胞,已被深入研究为多种疾病的治疗工具。为了增强MSCs的功效,使用逆转录病毒和慢病毒载体引入治疗基因。然而,严重不良事件(SAE)如肿瘤发生可通过插入诱变诱导。我们产生了编码野生型单纯疱疹病毒胸苷激酶(HSV-TK)基因和含有点突变的基因的慢病毒载体,所述点突变导致在残基168处的丙氨酸被替换为组氨酸(TK(A168H))并在MSC中转导表达(MSC-TK和MSC-TK(A168H))。编码TK(A168H)突变体的慢病毒载体的转导不改变增殖能力,中胚层分化潜能,或MSCs的表面抗原性。MSC-TK(A168H)细胞遗传稳定,如核型分析所示。MSC-TK(A168H)对更昔洛韦(GCV)的反应是最大抑制浓度(IC50)的一半,比MSC-TK低10倍。因为发现MSC-TK(A168H)细胞是非致瘤性的,U87-TK(A168H)皮下肿瘤被用作SAE样病症,我们评估了伐更昔洛韦(vGCV)的效果,GCV的口服前药。U87-TK(A168H)肿瘤被200mg/kgvGCV比U87-TK肿瘤更有效地消融。这些结果表明MSC-TK(A168H)细胞对于治疗用途而言似乎是临床前安全的。我们提出用HSV-TK(A168H)进行的遗传修饰通过消除SAE期间的移植细胞(如不受控制的细胞增殖)而使基于同种异体MSC的离体治疗更安全。
    Human mesenchymal stem cells (MSCs) are multipotent stem cells that have been intensively studied as therapeutic tools for a variety of disorders. To enhance the efficacy of MSCs, therapeutic genes are introduced using retroviral and lentiviral vectors. However, serious adverse events (SAEs) such as tumorigenesis can be induced by insertional mutagenesis. We generated lentiviral vectors encoding the wild-type herpes simplex virus thymidine kinase (HSV-TK) gene and a gene containing a point mutation that results in an alanine to histidine substitution at residue 168 (TK(A168H)) and transduced expression in MSCs (MSC-TK and MSC-TK(A168H)). Transduction of lentiviral vectors encoding the TK(A168H) mutant did not alter the proliferation capacity, mesodermal differentiation potential, or surface antigenicity of MSCs. The MSC-TK(A168H) cells were genetically stable, as shown by karyotyping. MSC-TK(A168H) responded to ganciclovir (GCV) with an half maximal inhibitory concentration (IC50) value 10-fold less than that of MSC-TK. Because MSC-TK(A168H) cells were found to be non-tumorigenic, a U87-TK(A168H) subcutaneous tumor was used as a SAE-like condition and we evaluated the effect of valganciclovir (vGCV), an oral prodrug for GCV. U87-TK(A168H) tumors were more efficiently ablated by 200 mg/kg vGCV than U87-TK tumors. These results indicate that MSC-TK(A168H) cells appear to be pre-clinically safe for therapeutic use. We propose that genetic modification with HSV-TK(A168H) makes allogeneic MSC-based ex vivo therapy safer by eliminating transplanted cells during SAEs such as uncontrolled cell proliferation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    BACKGROUND: Cell therapy as a promising therapeutic modality to treat cancer has been intensively studied for decades. However, the clinical trials have indicated that patients under T cell therapy may develop severe cytokine release syndrome resulting in hospitalization or even death. Furthermore, genetic modifications to promote proliferation and persistence of T cells could result in high numbers of long-lived engineered cells in patients after treatment.
    METHODS: We incorporated the pro-apoptotic truncated BH3 interacting-domain death agonist (tBID) with the mutant ecDHFR destabilizing domain to form a novel recombinant protein as the major component of an engineered tBID-based safety switch system, which would be unstable and quickly degraded in the absence of trimethoprim (TMP) but, upon TMP treatment, should become stabilized and allow tBID to induce cell death experimentally.
    RESULTS: The novel tBID-based safety switch could be regulated through a small molecule inducer, TMP, to control undesired toxicity or ablate the engineered cells as needed. We systematically compared and assessed several tBID-based safety switch constructs with the clinically validated safety switches, including human herpes simplex virus thymidine kinase (HSV-TK) and inducible Caspase 9 (iCasp9). With optimization, we were able to achieve significant killing potency in vitro in Jurkat or human primary T cells.
    CONCLUSIONS: We demonstrated that our engineered tBID-based safety switch was able to eliminate up to ~90% of transduced human primary T cells within 72 h after activation, providing an alternative switch system to manage safety concerns for cell therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    基于免疫细胞的疗法可以诱导有效的抗肿瘤作用,但也经常与严重的毒性有关。我们之前开发了一种基于PD-1的小分子调节的可逆性T细胞激活开关,以控制细胞免疫治疗产品的活性。这种化学调节的和SH2递送的抑制性尾部(CRASH-IT)开关依赖于开关SH2结构域与嵌合抗原受体或T细胞受体中磷酸化ITAM基序的非共价相互作用。经过这种互动,基于免疫受体酪氨酸的抑制基序/开关基序(ITIM/ITSM)包含存在于CRASH-IT开关中的PD-1结构域诱导T细胞信号传导的强烈抑制,和CRASH-IT介导的T细胞活性抑制可以通过小分子诱导的开关蛋白水解来逆转。为了开发改进的第二代开关系统,我们在这里分析了允许控制T细胞活性的免疫细胞受体对接和抑制性信号传导结构域的可能性空间.重要的是,这些分析表明,在原代人T细胞中最有效抑制抗原受体信号传导的抑制性结构域不是来自抑制性受体,如PD-1和BTLA,在T细胞中内源性表达,但包括含有来自骨髓细胞中存在的受体的抑制性结构域的ITIM/ITSM。此外,我们证明了抑制性结构域与抗原受体的物理接近对于有效抑制T细胞活化至关重要,仅使用SH2结构域直接与抗原受体中的ITAM基序相互作用的开关设计有效且可逆地抑制T细胞功能。这些数据证明了免疫细胞信号结构域的灵活和可互换的性质,并告知具有优越动态范围的合成接近开关系统的设计。
    Immune cell-based therapies can induce potent antitumor effects but are also often associated with severe toxicities. We previously developed a PD-1-based small molecule-regulated reversible T cell activation switch to control the activity of cellular immunotherapy products. This chemically regulated and SH2-delivered-inhibitory tail (CRASH-IT) switch relies on the noncovalent interaction of switch SH2 domains with phosphorylated ITAM motifs in either chimeric antigen receptors or T cell receptors. After this interaction, the immunoreceptor tyrosine-based inhibition motif/switch motif (ITIM/ITSM) containing PD-1 domain present in the CRASH-IT switch induces robust inhibition of T cell signaling, and CRASH-IT-mediated suppression of T cell activity can be reversed by small molecule-induced switch proteolysis. With the aim to develop improved second-generation switch systems, we here analyze the possibility space of both the immune cell receptor docking and inhibitory signaling domains that allow control over T cell activity. Importantly, these analyses demonstrate that the inhibitory domains that most potently suppress antigen receptor signaling in primary human T cells are not derived from inhibitory receptors, such as PD-1 and BTLA, that are endogenously expressed in T cells, but include ITIM/ITSM containing inhibitory domains derived from receptors present in myeloid cells. In addition, we demonstrate that physical proximity of the inhibitory domain to the antigen receptor is crucial to efficiently suppress T cell activation, as only switch designs that employ SH2 domains directly interacting with ITAM motifs in antigen receptors efficiently and reversibly inhibit T cell functionality. These data demonstrate the flexible and interchangeable nature of immune cell signaling domains, and inform the design of a synthetic proximity-based switch system with a superior dynamic range.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Induced pluripotent stem cell (iPSC)-derived cell products hold great promise as a potential cell source in personalized medicine. As concerns about the potential risk of graft-related severe adverse events, such as tumor formation from residual pluripotent cells, currently restrict their applicability, we established an optimized tool for therapeutic intervention that allows drug-controlled, specific and selective ablation of either iPSCs or the whole graft through genetic safety switches. To identify the best working system, different tools for genetic iPSC modification, promoters to express safety switches and different safety switches were combined. Suicide effects were slightly stronger when the suicide gene was delivered through lentiviral (LV) vectors compared to integration into the AAVS1 locus through TALEN technology. An optimized HSV-thymidine kinase and the inducible Caspase 9 both mediated drug-induced, efficient in vitro elimination of transgene-positive iPSCs. Choice of promoter allowed selective elimination of distinct populations within the graft: the hOct4 short response element restricted transgene expression to iPSCs, while the CAGs promoter ubiquitously drove expression in iPSCs and their progeny. Remarkably, both safety switches were able to prevent in vivo teratoma development and even effectively eliminated established teratomas formed by LV CAGs-transgenic iPSCs. These optimized tools to increase safety provide an important step towards clinical application of iPSC-derived transplants.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Case Reports
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号