adoptive cell transfer

过继细胞转移
  • 文章类型: Journal Article
    背景:嵌合抗原受体T(CAR-T)细胞疗法是一项创新技术,在临床试验中显示出可喜的结果。治疗基于修饰患者自身的T细胞以表达人工表面受体来特异性识别和攻击肿瘤细胞。
    目的:综合关于接受CAR-T细胞治疗的弥漫性大B细胞淋巴瘤患者细胞因子释放综合征发生率和治疗策略的现有证据。
    方法:这是一个系统的文献综述。搜索是在PubMed中进行的,Scopus,和Web科学数据库。本审查遵循了系统审查和荟萃分析(PRISMA)指南的首选报告项目。系统审查方案在国际前瞻性系统审查登记册(PROSPERO)数据库中注册,编号为CRD420223559258。
    结果:共纳入了19项研究,共1193名接受CAR-T细胞治疗的患者。在这些病人中,804(67%)发展了一定程度的细胞因子释放综合征。3级和4级细胞因子释放综合征的频率分别为10%和3%,分别。该综合征治疗中最常用的方案包括托珠单抗和/或糖皮质激素。
    结论:本综述的结果表明,在接受CAR-T细胞治疗的弥漫性大B细胞淋巴瘤患者中,细胞因子释放综合征的发生率很高,然而这些事件是可控的,支持该疗法在这些患者中是安全的结论。
    BACKGROUND: Chimeric antigen receptor T (CAR-T) cell therapy is an innovative technology that has shown promising results in clinical trials. Treatment is based on modifying the patient\'s own T cells to express artificial surface receptors to specifically recognize and attack the tumor cells.
    OBJECTIVE: To synthesize available evidence on the incidence and management strategies of cytokine release syndrome in patients with diffuse large B-cell lymphoma who received CAR-T cell therapy.
    METHODS: This is a systematic literature review. The search was conducted in the PubMed, Scopus, and Web of science databases. This review followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines. The systematic review protocol is registered in the International Prospective Register of Systematic Reviews (PROSPERO) database under number CRD42022359258.
    RESULTS: Nineteen studies were included with a total of 1193 patients who received CAR-T cell therapy. Of these patients, 804 (67%) developed some degree of cytokine release syndrome. The frequencies of Grade 3 and 4 cytokine release syndrome were 10% and 3%, respectively. The regimen most used in the management of the syndrome included tocilizumab and/or glucocorticoids.
    CONCLUSIONS: The results obtained in this review demonstrate high rates of cytokine release syndrome in patients with diffuse large B-cell lymphoma treated with CAR-T cell therapy, however these events are manageable, supporting the conclusion that this therapy is safe in these patients.
    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
    虽然现有的体外T细胞活化合成技术面临着诸如次优扩增率和低效率等挑战,人工抗原呈递细胞(aAPC)在增强T细胞治疗方面具有巨大前景.特别是,金纳米粒子(AuNPs),以其生物相容性而闻名,易于合成,和通用的表面化学,是用作纳米级aAPC的有力候选者。在这项研究中,我们开发了具有分支几何形状的尖刺AuNP,以将激活配体提供给原代人T细胞。尖峰AuNP的特殊结构增强了生物分子的装载能力,并通过共刺激配体和受体的多价结合显着提高了T细胞的活化。通过促进T细胞的更大多多克隆扩增,我们的尖峰AuNP优于现有系统,包括Dynabeads和可溶性激活剂,促进持续的细胞因子产生,并产生高功能的T细胞,减少疲惫。此外,尖峰AuNP有效激活和扩增CD19CAR-T细胞,同时使用比Dynabeads更少的效应细胞证明对靶细胞的体外细胞毒性增加。这项研究强调了尖峰AuNPs作为一种强大工具的潜力,为过继细胞疗法的应用带来新的机遇。
    While existing synthetic technologies for ex vivo T-cell activation face challenges like suboptimal expansion rates and low effectiveness, artificial antigen-presenting cells (aAPCs) hold great promise for enhanced T-cell based therapies. In particular, gold nanoparticles (AuNPs), known for their biocompatibility, ease of synthesis, and versatile surface chemistry, are strong candidates for use as nanoscale aAPCs. In this study, we developed spiky AuNPs with branched geometries to present activating ligands to primary human T-cells. The special structure of spiky AuNPs enhances biomolecule loading capacity and significantly improves T-cell activation through multivalent binding of costimulatory ligands and receptors. Our spiky AuNPs outperform existing systems including Dynabeads and soluble activators by promoting greater polyclonal expansion of T-cells, boosting sustained cytokine production, and generating highly functional T-cells with reduced exhaustion. In addition, spiky AuNPs effectively activate and expand CD19 CAR-T cells while demonstrating increased in vitro cytotoxicity against target cells using fewer effector cells than Dynabeads. This study underscores the potential of spiky AuNPs as a powerful tool, bringing new opportunities to adoptive cell therapy applications.
    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
    由于复杂的抗原呈递,抗原特异性T细胞活化不足会阻碍免疫治疗。此外,治疗性体内T细胞扩增受限于缓慢的扩增速率和有限的功能性。在这里,我们引入了一种称为抗原呈递细胞模拟融合蛋白(APC模拟物)的模型融合蛋白,旨在极大地模拟抗原呈递细胞的天然抗原呈递模式,并在体外和体内直接扩增T细胞。APC模拟物包含同源肽-人白细胞抗原(pHLA)复合物和共刺激标记CD80,它们是APC上的天然配体。在一次刺激之后,在不需要专门的抗原呈递细胞的情况下,与未处理组相比,APC模拟物导致抗原特异性T细胞的多克隆扩增增加约400倍。通过单细胞TCR测序(scTCR-seq)和单细胞RNA测序(scRNA-seq)的结合,我们在这些多克隆克隆型中鉴定出大约600倍的单克隆扩增克隆型.它还表现出在OT-1小鼠模型中证实的体内应用的适用性。此外,通过APC模拟物扩增的T细胞有效抑制过继性细胞转移(ACT)鼠模型中的肿瘤生长。这些发现为个性化治疗的多功能APC模拟平台铺平了道路,能够在体外和体内直接扩增多功能抗原特异性T细胞亚群。
    Inadequate antigen-specific T cells activation hampers immunotherapy due to complex antigen presentation. In addition, therapeutic in vivo T cell expansion is constrained by slow expansion rates and limited functionality. Herein, we introduce a model fusion protein termed antigen-presenting cell-mimic fusion protein (APC-mimic), designed to greatly mimicking the natural antigen presentation pattern of antigen-presenting cells and directly expand T cells both in vitro and in vivo. The APC-mimic comprises the cognate peptide-human leukocyte antigen (pHLA) complex and the co-stimulatory marker CD80, which are natural ligands on APCs. Following a single stimulation, APC-mimic leads to an approximately 400-fold increase in the polyclonal expansion of antigen-specific T cells compared with the untreated group in vitro without the requirement for specialized antigen-presenting cells. Through the combination of single-cell TCR sequencing (scTCR-seq) and single-cell RNA sequencing (scRNA-seq), we identify an approximately 600-fold monoclonal expansion clonotype among these polyclonal clonotypes. It also exhibits suitability for in vivo applications confirmed in the OT-1 mouse model. Furthermore, T cells expanded by APC-mimic effectively inhibits tumor growth in adoptive cell transfer (ACT) murine models. These findings pave the way for the versatile APC-mimic platform for personalized therapeutics, enabling direct expansion of polyfunctional antigen-specific T cell subsets in vitro and in vivo.
    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
    B细胞受体(BCR)转基因小鼠允许控制初始B细胞的初始靶标(抗原)特异性,并在活化后研究它们的性质。这里,我描述了如何将BCR转基因B细胞与过继细胞转移和免疫模型结合使用,以研究记忆B细胞的形成和再激活。
    B cell receptor (BCR) transgenic mice allow the control of the initial target (antigen) specificity of naïve B cells and to investigate their properties following activation. Here, I describe how BCR transgenic B cells can be used in combination with adoptive cell transfer and immunization models to study memory B cell formation and reactivation.
    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
    背景:移植的人胰岛组织的稀缺和需要免疫抑制药物来预防同种异体移植物的排斥反应,阻碍了通过胰岛移植治疗自身免疫性1型糖尿病(T1DM)。然而,过继转移的骨髓细胞(BMC)治疗有希望,这已经成为未来药物的有利途径。BMC有可能显著影响一系列疾病的替代和再生疗法。包括糖尿病,并且已经证明了抗糖尿病的作用。
    目的:本研究的主要目的是评估过继性转移的来自初始小鼠(nBMC)或糖尿病小鼠(dBMC)的骨髓细胞在治疗T1DM小鼠模型中的有效性。
    方法:雄性瑞士白化病小鼠饥饿16小时,然后以40mg/kg体重的剂量连续5天注射链脲佐菌素(STZ)以诱导T1DM。14天后,糖尿病小鼠分为四组。第一组作为糖尿病对照,用柠檬酸钠缓冲液治疗,而其他三组治疗两周,分别,胰岛素(皮下剂量为8U/kg/天),nBMC(静脉内剂量为1×106个细胞/小鼠/一次),和dBMC(以1×106个细胞/小鼠/一次的剂量静脉内)。
    结果:值得注意的是,对STZ诱导的T1DM小鼠施用过继转移的nBMC或过继转移的dBMC可显著改善血糖状况,伴随着血糖和糖化血红蛋白(HbA1C%)水平的显着降低,最终将对照小鼠的血清胰岛素水平恢复到初始状态。对STZ诱导的T1DM小鼠施用nBMC或dBMC导致血清中炎性细胞因子标志物水平显著降低,包括干扰素-γ(INF-γ),肿瘤坏死因子-α(TNF-α),肿瘤生长因子-β(TGF-β),白细胞介素-1β(L-1β),交织-4(IL-4),白细胞介素-6(IL-6),和白细胞介素-10(IL-10)。此外,STZ诱导的T1DM小鼠,当用nBMC或dBMC治疗时,总免疫球蛋白(Ig)水平显着上升。此外,胰岛细胞自身抗体(ICA)和胰岛素自身抗体(IAA)水平显著降低.此外,STZ诱导的T1DM小鼠血清中锌转运体8抗原蛋白(ZnT8)显著升高,胰岛抗原2蛋白(IA-2),和谷氨酸脱羧酶抗原蛋白(GAD)水平。有趣的是,在用nBMC或dBMC治疗的STZ诱导的T1DM小鼠中,施用nBMC或dBMC导致IA-2蛋白表达增强.此外,丙二醛(MDA)水平升高,而未治疗的STZ诱导的T1DM小鼠的过氧化氢酶(CAT)和超氧化物歧化酶(SOD)水平降低。然而,当向STZ诱导的T1DM小鼠施用nBMC或dBMC时,它对减少氧化应激有显著影响。这是通过降低血清中MDA的水平并增强酶抗氧化剂如CAT和SOD的活性来实现的。STZ诱导的T1DM小鼠肝脏酶ALT和AST水平显着升高,以及肌酐和尿素水平升高。考虑到肝脏和肾脏在代谢和排泄中的关键作用,这项研究进一步检查了对STZ诱导的T1DM小鼠施用nBMC或dBMC的效果。值得注意的是,这些细胞的施用减轻了观察到的效果。
    结论:本研究表明,在T1DM治疗中利用过继转移的nBMC或过继转移的dBMC导致血糖水平显著下降,可能归因于它们增强胰岛素分泌和改善胰岛性能的能力。此外,BMCs可能通过免疫调节对糖尿病小鼠的胰岛发挥有益作用,抗氧化剂,抗炎,和抗氧化应激特性。
    BACKGROUND: The scarcity of transplanted human islet tissue and the requirement for immunosuppressive drugs to prevent the rejection of allogeneic grafts have hindered the treatment of autoimmune type 1 diabetes mellitus (T1DM) through islet transplantation. However, there is hope in adoptively transferred bone marrow cells (BMCs) therapy, which has emerged as a propitious pathway for forthcoming medications. BMCs have the potential to significantly impact both replacement and regenerative therapies for a range of disorders, including diabetes mellitus, and have demonstrated anti-diabetic effects.
    OBJECTIVE: The main goal of this study is to evaluate the effectiveness of adoptively transferred bone marrow cells derived from either naïve mice (nBMCs) or diabetic mice (dBMCs) in treating a T1DM mice model.
    METHODS: Male Swiss albino mice were starved for 16 h and then injected with streptozotocin (STZ) at a dose of 40 mg/kg body weight for 5 consecutive days to induce T1DM. After 14 days, the diabetic mice were distributed into four groups. The first group served as a diabetic control treated with sodium citrate buffer, while the other three groups were treated for two weeks, respectively, with insulin (subcutaneously at a dose of 8 U/kg/day), nBMCs (intravenously at a dose of 1 × 106 cells/mouse/once), and dBMCs (intravenously at a dose of 1 × 106 cells/mouse/once).
    RESULTS: It is worth noting that administering adoptively transferred nBMCs or adoptively transferred dBMCs to STZ-induced T1DM mice resulted in a significant amelioration in glycemic condition, accompanied by a considerable reduction in the level of blood glucose and glycosylated hemoglobin % (HbA1C %), ultimately restoring serum insulin levels to their initial state in control mice. Administering nBMCs or dBMCs to STZ-induced T1DM mice led to a remarkable decrease in levels of inflammatory cytokine markers in the serum, including interferon-γ (INF-γ), tumor necrosis factor- α (TNF-α), tumor growth factor-β (TGF-β), interleukin-1 β (L-1β), interlekin-4 (IL-4), interleukin-6 (IL-6), and interleukin-10 (IL-10). Additionally, STZ-induced T1DM mice, when treated with nBMCs or dBMCs, experienced a notable rise in total immunoglobulin (Ig) level. Furthermore, there was a significant reduction in the levels of islet cell autoantibodies (ICA) and insulin autoantibodies (IAA). Furthermore, the serum of STZ-induced T1DM mice showed a significant increase in Zinc transporter 8 antigen protein (ZnT8), islet antigen 2 protein (IA-2), and glutamic acid decarboxylase antigen protein (GAD) levels. Interestingly, the administration of nBMCs or dBMCs resulted in a heightened expression of IA-2 protein in STZ-induced T1DM mice treated with nBMCs or dBMCs. Furthermore, the level of malondialdehyde (MDA) was increased, while the levels of catalase (CAT) and superoxide dismutase (SOD) were decreased in non-treated STZ-induced T1DM mice. However, when nBMCs or dBMCs were administered to STZ-induced T1DM mice, it had a significant impact on reducing oxidative stress. This was accomplished by reducing the levels of MDA in the serum and enhancing the activities of enzymatic antioxidants like CAT and SOD. STZ-induced T1DM mice displayed a significant elevation in the levels of liver enzymes ALT and AST, as well as heightened levels of creatinine and urea. Considering the crucial roles of the liver and kidney in metabolism and excretion, this research further examined the effects of administering nBMCs or dBMCs to STZ-induced T1DM mice. Notably, the administration of these cells alleviated the observed effects.
    CONCLUSIONS: The present study suggests that utilizing adoptively transferred nBMCs or adoptively transferred dBMCs in the treatment of T1DM led to noteworthy decreases in blood glucose levels, possibly attributed to their capacity to enhance insulin secretion and improve the performance of pancreatic islets. Additionally, BMCs may exert their beneficial effects on the pancreatic islets of diabetic mice through their immunomodulatory, antioxidant, anti-inflammatory, and anti-oxidative stress properties.
    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
    免疫检查点(IC)阻断和肿瘤特异性T细胞(ACT)的过继转移是治疗转移性黑色素瘤的两种主要策略。它们的组合可以增强抑制性肿瘤微环境中的T细胞激活,但与全身注射IC阻滞剂相关的自身免疫不良反应在此策略中持续存在.IC表达缺陷的肿瘤反应性T细胞的ACT将克服这个问题。为此,PD-1和TIGIT似乎是相关的候选人,因为它们在高度肿瘤反应性淋巴细胞上的共表达限制了它们在肿瘤微环境中的治疗功效,NT.我们的研究比较了PDCD1或TIGIT基因缺失对黑色素瘤特异性T淋巴细胞的抗肿瘤特性和T细胞适应性的影响。转录组学分析显示PD-1KOT细胞中细胞周期相关基因的下调,与生物学观察一致,而增殖途径保留在TIGITKOT细胞中。功能分析表明,PD-1KO和TIGITKOT细胞在体外和使用免疫缺陷小鼠的临床前黑色素瘤模型中表现出比其野生型对应物更好的抗肿瘤反应性。有趣的是,看来TIGITKOT细胞在体内更有效地抑制肿瘤细胞增殖,在肿瘤内的存留时间比PD-1KOT细胞长,与TIGIT缺失对T细胞适应性没有影响一致。一起来看,这些结果表明TIGIT缺失,超过PD-1删除,黑色素瘤特异性T细胞是未来免疫治疗策略的一个令人信服的选择.
    Immune checkpoint (IC) blockade and adoptive transfer of tumor-specific T-cells (ACT) are two major strategies to treat metastatic melanoma. Their combination can potentiate T-cell activation in the suppressive tumor microenvironment, but the autoimmune adverse effects associated with systemic injection of IC blockers persist with this strategy. ACT of tumor-reactive T-cells defective for IC expression would overcome this issue. For this purpose, PD-1 and TIGIT appear to be relevant candidates, because their co-expression on highly tumor-reactive lymphocytes limits their therapeutic efficacy within the tumor microenvironme,nt. Our study compares the consequences of PDCD1 or TIGIT genetic deletion on anti-tumor properties and T-cell fitness of melanoma-specific T lymphocytes. Transcriptomic analyses revealed down-regulation of cell cycle-related genes in PD-1KO T-cells, consistent with biological observations, whereas proliferative pathways were preserved in TIGITKO T-cells. Functional analyses showed that PD-1KO and TIGITKO T-cells displayed superior antitumor reactivity than their wild-type counterpart in vitro and in a preclinical melanoma model using immunodeficient mice. Interestingly, it appears that TIGITKO T-cells were more effective at inhibiting tumor cell proliferation in vivo, and persist longer within tumors than PD-1KO T-cells, consistent with the absence of impact of TIGIT deletion on T-cell fitness. Taken together, these results suggest that TIGIT deletion, over PD-1 deletion, in melanoma-specific T-cells is a compelling option for future immunotherapeutic strategies.
    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
    近年来,免疫疗法行业的到来带来了提供变革的可能性,耐用,以及各种恶性肿瘤的潜在治愈结果。然而,进一步的研究表明,有许多问题显着降低了免疫疗法的有效性,尤其是实体瘤。首先,这些问题与肿瘤及其微环境的保护机制有关。目前,主要努力集中在通过使用不同的过继细胞疗法变体和基因工程构建体的修饰来克服保护机制。此外,需要复杂的劳动力来开发和实施这些治疗方法。为了克服这些重大挑战,创新的策略和方法是必要的工程更强大的免疫疗法的变化,提高抗肿瘤活性和降低毒性.在这次审查中,我们讨论了最近的免疫疗法创新,旨在提高实体瘤的临床疗效,以及克服各种免疫疗法局限性的策略。
    In recent years, the arrival of the immunotherapy industry has introduced the possibility of providing transformative, durable, and potentially curative outcomes for various forms of malignancies. However, further research has shown that there are a number of issues that significantly reduce the effectiveness of immunotherapy, especially in solid tumors. First of all, these problems are related to the protective mechanisms of the tumor and its microenvironment. Currently, major efforts are focused on overcoming protective mechanisms by using different adoptive cell therapy variants and modifications of genetically engineered constructs. In addition, a complex workforce is required to develop and implement these treatments. To overcome these significant challenges, innovative strategies and approaches are necessary to engineer more powerful variations of immunotherapy with improved antitumor activity and decreased toxicity. In this review, we discuss recent innovations in immunotherapy aimed at improving clinical efficacy in solid tumors, as well as strategies to overcome the limitations of various immunotherapies.
    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
    蛋白酶体产生大部分呈递在MHCI类分子上的肽。已显示蛋白酶体的切割模式通过蛋白酶体激活剂(PA)28αβ(PA28αβ)改变。特别是,已经报道了几种免疫原性肽是PA28αβ依赖性的。相比之下,我们没有观察到PA28αβ对不同主要组织相容性复合物(MHC)I类配体的产生有重大影响。感染淋巴细胞脉络膜脑膜炎病毒(LCMV)或牛痘病毒的PA28αβ敲除小鼠显示出正常的分化簇(CD)8应答和病毒清除。然而,我们观察到野生型细胞过继转移到PA28αβ基因敲除小鼠中导致移植物排斥,但反之亦然。耗竭实验表明,观察到的排斥是由CD8+细胞毒性T细胞介导的。这些数据表明PA28αβ可能参与胸腺中CD8+T细胞库的发育。一起来看,我们的数据表明,PA28αβ是决定T细胞选择的关键因素,因此,影响移植物接受。
    The proteasome generates the majority of peptides presented on MHC class I molecules. The cleavage pattern of the proteasome has been shown to be changed via the proteasome activator (PA)28 alpha beta (PA28αβ). In particular, several immunogenic peptides have been reported to be PA28αβ-dependent. In contrast, we did not observe a major impact of PA28αβ on the generation of different major histocompatibility complex (MHC) classI ligands. PA28αβ-knockout mice infected with the lymphocytic choriomeningitis virus (LCMV) or vaccinia virus showed a normal cluster of differentiation (CD) 8 response and viral clearance. However, we observed that the adoptive transfer of wild-type cells into PA28αβ-knockout mice led to graft rejection, but not vice versa. Depletion experiments showed that the observed rejection was mediated by CD8+ cytotoxic T cells. These data indicate that PA28αβ might be involved in the development of the CD8+ T cell repertoire in the thymus. Taken together, our data suggest that PA28αβ is a crucial factor determining T cell selection and, therefore, impacts graft acceptance.
    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
    多发性骨髓瘤(MM)仍然无法治愈,尽管出现了嵌合抗原受体(CAR)-T细胞疗法。这种未实现的潜力可归因于两个未解决的问题:缺乏合适的CAR目标和格式。相对于前者,目标应该是高表达和不愿意脱落;归因于CS1抗原的两个特征。此外,传统的CAR依赖于scFvs进行抗原识别,然而,这隐瞒了不利之处,主要是由这种格式的内在不稳定性造成的。已提出VHH作为有效的scFv替代物。因此,我们打算开发基于VHH的CAR-T细胞,靶向CS1,并鉴定诱导最佳CAR-T细胞激活的VHH以及实现此目的所需的VHH参数。
    生成了CS1特定的VHH,识别和充分表征,在体外和体内。接下来,它们被整合到第二代CAR中,这些CAR的抗原结合部分仅不同.用不同的VHH-CAR慢病毒转导报告T细胞系,并且并排评估CAR-T细胞活化动力学。亲和力,细胞结合能力,表位位置,体内行为,绑定距离,研究了CAR-T:MM细胞相互作用对的方向作为CAR-T细胞活化的预测参数。
    我们的数据表明,VHHs对其靶抗原的亲和力对其体内肿瘤示踪能力具有相对预测性,因为在MM的体内模型中,肿瘤摄取通常随着亲和力的降低而降低。这并不适用于他们的CAR-T细胞激活潜力,因为一些中间亲和力结合VHHs被证明非常有效,而一些较高亲和力的VHH未能诱导相等水平的T细胞活化。这不能归因于细胞结合能力,体内VHH行为,表位位置,细胞间距离或结合方向。因此,没有一个研究的参数证明对CAR-T细胞活化的程度具有显著的预测价值。
    我们使用针对高度相关的MM抗原CS1的VHH文库深入了解了CAR背景下VHH的预测参数。由于研究的VHH参数都没有预测价值,定义最佳CAR-T细胞激活的VHHs仍然与偶然性有关。这些发现强调了筛选多个候选人的重要性。
    UNASSIGNED: Multiple myeloma (MM) remains incurable, despite the advent of chimeric antigen receptor (CAR)-T cell therapy. This unfulfilled potential can be attributed to two untackled issues: the lack of suitable CAR targets and formats. In relation to the former, the target should be highly expressed and reluctant to shedding; two characteristics that are attributed to the CS1-antigen. Furthermore, conventional CARs rely on scFvs for antigen recognition, yet this withholds disadvantages, mainly caused by the intrinsic instability of this format. VHHs have been proposed as valid scFv alternatives. We therefore intended to develop VHH-based CAR-T cells, targeting CS1, and to identify VHHs that induce optimal CAR-T cell activation together with the VHH parameters required to achieve this.
    UNASSIGNED: CS1-specific VHHs were generated, identified and fully characterized, in vitro and in vivo. Next, they were incorporated into second-generation CARs that only differ in their antigen-binding moiety. Reporter T-cell lines were lentivirally transduced with the different VHH-CARs and CAR-T cell activation kinetics were evaluated side-by-side. Affinity, cell-binding capacity, epitope location, in vivo behavior, binding distance, and orientation of the CAR-T:MM cell interaction pair were investigated as predictive parameters for CAR-T cell activation.
    UNASSIGNED: Our data show that the VHHs affinity for its target antigen is relatively predictive for its in vivo tumor-tracing capacity, as tumor uptake generally decreased with decreasing affinity in an in vivo model of MM. This does not hold true for their CAR-T cell activation potential, as some intermediate affinity-binding VHHs proved surprisingly potent, while some higher affinity VHHs failed to induce equal levels of T-cell activation. This could not be attributed to cell-binding capacity, in vivo VHH behavior, epitope location, cell-to-cell distance or binding orientation. Hence, none of the investigated parameters proved to have significant predictive value for the extent of CAR-T cell activation.
    UNASSIGNED: We gained insight into the predictive parameters of VHHs in the CAR-context using a VHH library against CS1, a highly relevant MM antigen. As none of the studied VHH parameters had predictive value, defining VHHs for optimal CAR-T cell activation remains bound to serendipity. These findings highlight the importance of screening multiple candidates.
    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
    人乳头瘤病毒16型(HPV16)引起很大一部分生殖器和口咽癌。为了保持转换后的状态,肿瘤细胞必须连续合成E6和E7病毒癌蛋白,这使得它们成为肿瘤特异性抗原。的确,针对它们的特异性T细胞反应已经得到了很好的证明,并且经工程改造以表达识别E6或E7表位的T细胞受体(TCR)的CD8+T细胞已经在临床研究中进行了测试,结果有希望。但临床成功有限。使用来自健康供体外周血的CD8+T细胞,我们已经鉴定了两个新的与未探索的E618-26表位反应的TCR。这些TCR对E618-26-HLA-A*02:01呈递肿瘤细胞显示有限的独立细胞毒性。然而,靶向L1CAM的单信号传导域嵌合抗原受体(ssdCAR),一种在HPV16诱导的癌症中经常过表达的细胞粘附蛋白,提示协同作用,显着增强NK-92/CD3/CD8细胞装甲TCR和ssdCAR的细胞毒性能力,当两种受体同时参与各自的目标,如通过2-D和3-D共培养物的活显微镜显示的。因此,来自健康供体CD8+T细胞库的病毒特异性TCR可以与合适的ssdCAR组合,以增强效应细胞的细胞毒性能力,间接地,他们的特异性。
    The human papillomavirus type 16 (HPV16) causes a large fraction of genital and oropharyngeal carcinomas. To maintain the transformed state, the tumor cells must continuously synthesize the E6 and E7 viral oncoproteins, which makes them tumor-specific antigens. Indeed, specific T cell responses against them have been well documented and CD8+ T cells engineered to express T cell receptors (TCRs) that recognize epitopes of E6 or E7 have been tested in clinical studies with promising results, yet with limited clinical success. Using CD8+ T cells from peripheral blood of healthy donors, we have identified two novel TCRs reactive to an unexplored E618-26 epitope. These TCRs showed limited standalone cytotoxicity against E618-26-HLA-A*02:01-presenting tumor cells. However, a single-signaling domain chimeric antigen receptor (ssdCAR) targeting L1CAM, a cell adhesion protein frequently overexpressed in HPV16-induced cancer, prompted a synergistic effect that significantly enhanced the cytotoxic capacity of NK-92/CD3/CD8 cells armored with both TCR and ssdCAR when both receptors simultaneously engaged their respective targets, as shown by live microscopy of 2-D and 3-D co-cultures. Thus, virus-specific TCRs from the CD8+ T cell repertoire of healthy donors can be combined with a suitable ssdCAR to enhance the cytotoxic capacity of the effector cells and, indirectly, their specificity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号