T cell engager

T 细胞衔接器
  • 文章类型: Journal Article
    针对非适应性免疫细胞的治疗策略目前正在临床开发中。γδT细胞是T细胞的小亚型(占总T细胞的1-10%),无需抗原呈递机制即可介导其效应子功能。并与先天细胞共享功能特性。在不同的γδT亚型中,抗Vγ9Vδ2T抗体在早期临床研究中报告了临床疗效的迹象.在这篇综述中,我们描述了这种非常规T细胞亚型的生物学,并提供了激活这些细胞的新型抗体的作用机制的见解。我们将专注于靶向BTN3A配体和双特异性γδT细胞衔接剂的抗体。我们将详细回顾这些策略的优势,包括克服检查点抑制剂耐药机制的潜力,或与激活经典T细胞的药物相比更充分的安全性。在人类的第一次研究中确定的限制和克服这些限制的策略将被修改和讨论。最后,将为未来的临床发展提出临床选择。
    Therapeutic strategies targeting non-adaptive immune cells are currently in clinical development. γδT cells are a small subtype of T cells (1-10% of total T cells) that mediate their effector function without the necessity of the antigen presenting machinery, and also share functional properties with innate cells. Among the different γδT subtypes, antibodies against Vγ9Vδ2T have reported signs of clinical efficacy in early clinical studies. In this review we describe the biology of this subtype of non-conventional T cells and provide insights into the mechanism of action of novel antibodies that activate these cells. We will focus on antibodies targeting the BTN3A ligand and bi-specific γδT cell engagers. We will review in detail the advantages of these strategies including the potential for overcoming mechanisms of resistance to check point inhibitors, or the much more adequate safety profile compared with agents activating classical T cells. Limitations identified during the first studies in humans and strategies to overcome them will be revised and discussed. Finally, clinical options for future clinical development will be suggested.
    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
    背景:白细胞介素-1受体辅助蛋白(IL1RAP)在急性髓性白血病(AML)大母细胞和白血病干细胞(LSCs)上高表达,但不是在正常造血干细胞(HSC),提供瞄准和消除疾病的机会,同时保留正常的造血功能。在这里,我们报道了新型抗IL1RAP/CD3T细胞接合剂(TCE)BIF002在AML中的活性。
    方法:通过光电定位和单细胞测序,从从免疫小鼠收集的CD138+B细胞中分离IL1RAP抗体。产生并表征单个小鼠单克隆抗体(mAb),我们从其中产生BIF002,一种使用Fab臂交换的抗人IL1RAP/CD3TCE。在人IgG1Fc中引入突变以减少FcγR结合。使用多种细胞系和患者来源的AML样品在体外和体内表征BIF002的抗白血病活性。
    结果:发现IL1RAP在大多数人AML细胞系和原代母细胞中高表达,包括从头和复发/难治性(R/R)白血病患者的CD34+LSC富集亚群,但不是正常的HSC。在来自健康供体和IL1RAPhighAML细胞系和原代母细胞的T细胞的共培养中,BIF002在亚纳摩尔浓度下诱导剂量和效应物对靶(E:T)比率依赖性T细胞活化和白血病细胞裂解。BIF002与人T细胞一起静脉内给药导致白血病细胞的消耗,并显著延长IL1RAPhighMOLM13或AML患者来源的异种移植物的存活,没有脱靶副作用,与对照组相比。值得注意的是,BiF002有效地重定向T细胞以消除LSCs,与来自载体-(中位生存期:26天;p=0.0004)或同种型对照抗体+T细胞治疗的供体(26天;p=0.0002)的骨髓(BM)的受体相比,来自BIF002+T细胞治疗的供体(中位生存期未达到;全部存活>200天)的骨髓(BM)的次级受体没有发生疾病。
    结论:新型抗IL1RAP/CD3TCE,BIF002根除LSCs并显著延长AML异种移植物的存活时间,代表一个有希望的人,AML的新疗法。
    BACKGROUND: The interleukin-1 receptor accessory protein (IL1RAP) is highly expressed on acute myeloid leukemia (AML) bulk blasts and leukemic stem cells (LSCs), but not on normal hematopoietic stem cells (HSCs), providing an opportunity to target and eliminate the disease, while sparing normal hematopoiesis. Herein, we report the activity of BIF002, a novel anti-IL1RAP/CD3 T cell engager (TCE) in AML.
    METHODS: Antibodies to IL1RAP were isolated from CD138+ B cells collected from the immunized mice by optoelectric positioning and single cell sequencing. Individual mouse monoclonal antibodies (mAbs) were produced and characterized, from which we generated BIF002, an anti-human IL1RAP/CD3 TCE using Fab arm exchange. Mutations in human IgG1 Fc were introduced to reduce FcγR binding. The antileukemic activity of BIF002 was characterized in vitro and in vivo using multiple cell lines and patient derived AML samples.
    RESULTS: IL1RAP was found to be highly expressed on most human AML cell lines and primary blasts, including CD34+ LSC-enriched subpopulation from patients with both de novo and relapsed/refractory (R/R) leukemia, but not on normal HSCs. In co-culture of T cells from healthy donors and IL1RAPhigh AML cell lines and primary blasts, BIF002 induced dose- and effector-to-target (E:T) ratio-dependent T cell activation and leukemic cell lysis at subnanomolar concentrations. BIF002 administered intravenously along with human T cells led to depletion of leukemic cells, and significantly prolonged survival of IL1RAPhigh MOLM13 or AML patient-derived xenografts with no off-target side effects, compared to controls. Of note, BiF002 effectively redirects T cells to eliminate LSCs, as evidenced by the absence of disease initiation in secondary recipients of bone marrow (BM) from BIF002+T cells-treated donors (median survival not reached; all survived > 200 days) compared with recipients of BM from vehicle- (median survival: 26 days; p = 0.0004) or isotype control antibody+T cells-treated donors (26 days; p = 0.0002).
    CONCLUSIONS: The novel anti-IL1RAP/CD3 TCE, BIF002, eradicates LSCs and significantly prolongs survival of AML xenografts, representing a promising, novel treatment for AML.
    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
    T细胞重定向疗法(TCRTs),例如CAR-或TCRT细胞和T细胞衔接者,已经成为一种非常有效的治疗方式,特别是在B和浆细胞恶性肿瘤环境中。然而,许多患者无法获得深刻而持久的反应;而缺乏真正独特的肿瘤抗原,和并发的靶/外肿瘤毒性,阻碍了许多其他癌症的TCRTs的发展。在这次审查中,我们讨论了血液恶性肿瘤TCRT靶点的最新进展,以及旨在解决这些问题的新目标策略,和其他,挑战。
    T cell-redirecting therapies (TCRTs), such as chimeric antigen receptor (CAR) or T cell receptor (TCR) T cells and T cell engagers, have emerged as a highly effective treatment modality, particularly in the B and plasma cell-malignancy setting. However, many patients fail to achieve deep and durable responses; while the lack of truly unique tumor antigens, and concurrent on-target/off-tumor toxicities, have hindered the development of TCRTs for many other cancers. In this review, we discuss the recent developments in TCRT targets for hematological malignancies, as well as novel targeting strategies that aim to address these, and other, challenges.
    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
    双特异性抗体是治疗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。我们表明,表达片段可结晶结构域缺陷型抗CD19×抗CD3(blinatumomab)或抗CD33×抗CD3双特异性抗体的人浆细胞可介导T细胞活化并直接杀死B急性淋巴细胞白血病或急性髓性白血病细胞系。我们证明了自我表达抗原的敲除,CD19促进浆细胞的抗CD19双特异性分泌并防止自我靶向。分泌抗CD19双特异性抗体的浆细胞在与自体T细胞共移植的免疫缺陷小鼠中引起急性淋巴细胞白血病患者来源的异种移植物的体内控制。在这些研究中,我们发现,工程浆细胞引起的白血病控制与CD19靶向嵌合抗原受体表达T细胞相似.最后,在细胞递送和肿瘤根除后1个月,血清中抗CD19双特异性药物的稳态浓度与在接受博纳吐单抗稳态输注治疗的患者中观察到的浓度相当.这些发现支持ePCs作为治疗急性白血病的持久递送系统的进一步发展。和潜在的其他癌症。
    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.
    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
    对于抗药物抗体(ADA)对相关暴露的影响敏感的适当设计的药代动力学(PK)测定是了解ADA的中和潜力的替代策略。然而,缺少有关如何开发此类PK测定以及如何确认ADA对暴露的功能性影响的指南。这里,T细胞接合双特异性抗体的PK测定,Cibisatamab,是根据其作用机制(MoA)开发的。使用关键的单克隆抗独特型(抗ID)抗体阳性对照作为ADA替代,对暴露的影响进行了临床前评估.在I期临床试验(NCT02324257)中,初始数据表明,ADA和PK测定的组合用于ADA反应与Cibisatamab暴露的相关性。为了了解患者来源的ADAs对药物活性的中和潜力,先进的ADA表征已经进行。评估了ADA与药物抗体结构域的结构结合分析及其对靶向的影响。为此,我们确定了相关患者ADA结合特征,并将其与特异性单克隆抗ID抗体阳性对照进行了比较.靶标结合抑制的可比较结果和对暴露的类似影响表明,在患者中观察到的Cmax和Ctrugh水平的降低是由ADAs的中和潜力引起的,并且允许ADA响应与暴露损失之间的相关性。因此,所描述的研究为开发适当设计的双特异性抗体PK测定提供了重要的功能方面,作为了解中和ADA对暴露影响的替代选择.
    An appropriately designed pharmacokinetic (PK) assay that is sensitive for anti-drug antibody (ADA) impact on relevant exposure is an alternative strategy to understand the neutralizing potential of ADAs. However, guidance on how to develop such PK assays and how to confirm the functional ADA impact on exposure is missing. Here, the PK assay of a T-cell-engaging bispecific antibody, cibisatamab, was developed based on its mechanism of action (MoA). Using critical monoclonal anti-idiotypic (anti-ID) antibody positive controls as ADA surrogates, the impact on exposure was evaluated pre-clinically. In a phase I clinical trial (NCT02324257), initial data suggest that the combination of ADA and PK assays for correlation of the ADA response with cibisatamab exposure. To understand the neutralizing potential of patient-derived ADAs on drug activity, advanced ADA characterization has been performed. Structural binding analysis of ADAs to antibody domains of the drug and its impact on targeting were assessed. For this purpose, relevant patient ADA binding features were identified and compared with the specific monoclonal anti-ID antibody-positive controls. Comparable results of target binding inhibition and similar impacts on exposure suggest that the observed reduction of Cmax and Ctrough levels in patients is caused by the neutralizing potential of ADAs and allows a correlation between ADA response and loss of exposure. Therefore, the described study provides important functional aspects for the development of an appropriately designed PK assay for bispecific antibodies as an alternative option towards understanding the neutralizing ADA impact on exposure.
    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
    在过去的十年里,目前正在开发免疫疗法,以增强免疫系统消除肿瘤细胞的能力。被称为T细胞衔接剂(TCE)的双特异性抗体在这种追求中提出了有吸引力的策略。TCEs旨在引导细胞毒性T细胞朝向肿瘤细胞,从而诱导强激活和随后的肿瘤细胞裂解。在这项研究中,我们研究了不同TCE对常规α-β(αβ)T细胞和非常规γδ(γδ)T细胞的活性。使用靶向肿瘤相关抗原CEACAM5(CEA)的骆驼单域抗体(VHHs)构建TCEs,以及T细胞受体链或CD3结构域。我们表明,当用CD3xCEATCE刺激时,Vγ9Vδ2T细胞比αβT细胞显示出更强的体外抗肿瘤活性。此外,限制新鲜人外周血T细胞对Vγ9Vδ2T细胞的激活限制了原肿瘤因子和促炎细胞因子的产生,通常与患者的毒性有关。一起来看,我们的发现提供了进一步的见解,即γδT细胞特异性TCEs有望作为特异性,有效,和潜在安全的分子来改善抗肿瘤免疫疗法。
    In the last decade, there has been a surge in developing immunotherapies to enhance the immune system\'s ability to eliminate tumor cells. Bispecific antibodies known as T cell engagers (TCEs) present an attractive strategy in this pursuit. TCEs aim to guide cytotoxic T cells toward tumor cells, thereby inducing a strong activation and subsequent tumor cell lysis. In this study, we investigated the activity of different TCEs on both conventional alpha-beta (αβ) T cells and unconventional gamma delta (γδ) T cells. TCEs were built using camelid single-domain antibodies (VHHs) targeting the tumor-associated antigen CEACAM5 (CEA), together with T cell receptor chains or a CD3 domain. We show that Vγ9Vδ2 T cells display stronger in vitro antitumor activity than αβ T cells when stimulated with a CD3xCEA TCE. Furthermore, restricting the activation of fresh human peripheral T cells to Vγ9Vδ2 T cells limited the production of protumor factors and proinflammatory cytokines, commonly associated with toxicity in patients. Taken together, our findings provide further insights that γδ T cell-specific TCEs hold promise as specific, effective, and potentially safe molecules to improve antitumor immunotherapies.
    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
    目前多发性骨髓瘤(MM)的治疗策略非常有效,但大多数患者发展复发/难治性疾病(RRMM)。抗CD38/CD3xCD28三特异性抗体SAR442257靶向MM细胞上的CD38和CD28并共刺激T细胞(TC)上的CD3和CD28。我们评估了不同的关键方面,如MM细胞和T细胞亲和力相互作用,肿瘤杀伤,和三个不同的RRMM患者队列中药物效力的生物标志物。我们发现,与新诊断的MM(NDMM)患者(19%)相比,显著更高比例的RRMM患者(86%)表现出CD28的异常共表达。此外,SAR442257介导的TC激活显著增高,与所有复发队列的双特异性功能敲除对照相比,导致MM杀伤增强(Pearson'sr=0.7)。最后,与其他队列相比,抗CD38治疗难治性患者的TGF-β水平较高(高达20倍).这可以限制SAR442257的活性。Vactoserib,TGF-β抑制剂,在这些实验中能够减轻这种影响并恢复对SAR442257的敏感性。总之,SAR442257具有通过共同靶向MM上的CD38和CD28以及T细胞上的CD3/CD28来增强TC细胞毒性的高潜力。
    Current treatment strategies for multiple myeloma (MM) are highly effective, but most patients develop relapsed/refractory disease (RRMM). The anti-CD38/CD3xCD28 trispecific antibody SAR442257 targets CD38 and CD28 on MM cells and co-stimulates CD3 and CD28 on T cells (TCs). We evaluated different key aspects such as MM cells and T cells avidity interaction, tumor killing, and biomarkers for drug potency in three distinct cohorts of RRMM patients. We found that a significantly higher proportion of RRMM patients (86%) exhibited aberrant co-expression of CD28 compared to newly diagnosed MM (NDMM) patients (19%). Furthermore, SAR442257 mediated significantly higher TC activation, resulting in enhanced MM killing compared to bispecific functional knockout controls for all relapse cohorts (Pearson\'s r = 0.7). Finally, patients refractory to anti-CD38 therapy had higher levels of TGF-β (up to 20-fold) compared to other cohorts. This can limit the activity of SAR442257. Vactoserib, a TGF-β inhibitor, was able to mitigate this effect and restore sensitivity to SAR442257 in these experiments. In conclusion, SAR442257 has high potential for enhancing TC cytotoxicity by co-targeting CD38 and CD28 on MM and CD3/CD28 on T cells.
    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
    肉瘤是难以治疗的罕见且异质的恶性肿瘤。诊断为肉瘤的患者中约有50%发展为转移性疾病,迄今为止的治疗选择非常有限。据报道,跨膜蛋白B7-H3在各种恶性肿瘤中表达,包括不同的肉瘤亚型.在一些癌症实体中,B7-H3表达与不良预后相关。反过来,B7-H3被认为是免疫治疗方法的有希望的靶标。我们在这里报道了基于IgG的B7-H3xCD3双特异性抗体的临床前特征,称为CC-3,用于治疗不同的肉瘤亚型。我们发现B7-H3在所有测试的肉瘤细胞上表达,并且在肉瘤患者上的表达与无进展和总体生存率降低相关。发现CC-3引起针对多种肉瘤亚型的强烈T细胞反应,导致显著的激活,细胞因子和效应分子的释放。此外,CC-3促进T细胞增殖和分化,导致记忆T细胞亚群的产生。最后,CC-3以靶细胞受限的方式诱导有效的靶细胞裂解。基于这些结果,目前正在准备一项评估CC-3在软组织肉瘤中的临床试验.
    Sarcomas are rare and heterogeneous malignancies that are difficult to treat. Approximately 50% of patients diagnosed with sarcoma develop metastatic disease with so far very limited treatment options. The transmembrane protein B7-H3 reportedly is expressed in various malignancies, including different sarcoma subtypes. In several cancer entities B7-H3 expression is associated with poor prognosis. In turn, B7-H3 is considered a promising target for immunotherapeutic approaches. We here report on the preclinical characterization of a B7-H3xCD3 bispecific antibody in an IgG-based format, termed CC-3, for treatment of different sarcoma subtypes. We found B7-H3 to be expressed on all sarcoma cells tested and expression on sarcoma patients correlated with decreased progression-free and overall survival. CC-3 was found to elicit robust T cell responses against multiple sarcoma subtypes, resulting in significant activation, release of cytokines and effector molecules. In addition, CC-3 promoted T cell proliferation and differentiation, resulting in the generation of memory T cell subsets. Finally, CC-3 induced potent target cell lysis in a target cell restricted manner. Based on these results, a clinical trial evaluating CC-3 in soft tissue sarcoma is currently in preparation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    系统性硬化症,严重的炎症性自身免疫性疾病,通过炎症的潜在机制与癌症有着共同的线索。这种炎症环境不仅驱动自身免疫性疾病的免疫调节失调特征,而且在癌症的发病机理中起关键作用。在涉及的细胞成分中,B细胞已经成为血液肿瘤和自身免疫性疾病的关键参与者,导致系统性硬化症的免疫失调和持续的组织纤维化,以及肿瘤进展和癌症中的免疫逃避。因此,靶向B细胞的新型治疗策略在这两种情况下都有希望。最近对CD19CART细胞在严重系统性硬化症患者中的探索显示出巨大的潜力,但也引入了与病毒载体相关的可能风险和缺点,延长CART细胞的持久性,漫长的生产时间表,高成本,以及对患者进行器官毒性和损害生育能力的化疗的必要性。鉴于这些挑战,替代的CD19消耗方法对于管理严重的系统性自身免疫性疾病非常感兴趣.这里,我们介绍了blinatumomab的开创性用途,在进行性,严重的系统性硬化症,为此类具有挑战性的案件提供了一个有希望的替代方案。
    Systemic sclerosis, a severe inflammatory autoimmune disease, shares a common thread with cancer through the underlying mechanism of inflammation. This inflammatory milieu not only drives the immune dysregulation characteristic of autoimmune diseases but also plays a pivotal role in the pathogenesis of cancer. Among the cellular components involved, B cells have emerged as key players in hematologic tumor and autoimmune disease, contributing to immune dysregulation and persistent tissue fibrosis in systemic sclerosis, as well as tumor progression and immune evasion in cancer. Consequently, novel therapeutic strategies targeting B cells hold promise in both conditions. Recent exploration of CD19 CAR T cells in severe systemic sclerosis patients has shown great potential, but also introduced possible risks and drawbacks associated with viral vectors, prolonged CAR T cell persistence, lengthy production timelines, high costs, and the necessity of conditioning patients with organotoxic and fertility-damaging chemotherapy. Given these challenges, alternative CD19-depleting approaches are of high interest for managing severe systemic autoimmune diseases. Here, we present the pioneering use of blinatumomab, a bispecific anti-CD3/anti-CD19 T cell engager in a patient with progressive, severe systemic sclerosis, offering a promising alternative for such challenging cases.
    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(CAR)T细胞疗法在实体瘤治疗中由于免疫抑制性肿瘤微环境和次优T细胞持久性而受阻。当前的策略没有解决微环境中的营养竞争。因此,我们提出了一种使用肌苷作为替代燃料的代谢加油方法。CART细胞被工程化以表达膜结合的CD26和胞质腺苷脱氨酶1(ADA1),将腺苷转化为肌苷。自分泌的ADA1在CD3/CD26刺激激活CART细胞,改善迁移和对转化生长因子β1抑制的抗性。ADA1与抗CD3scFv的融合进一步促进肌苷产生并使肿瘤细胞摄食最小化。在肝细胞癌和非小细胞肺癌的小鼠模型中,与未修饰的CART细胞相比,代谢加油的CART细胞表现出优异的肿瘤减少。总的来说,我们的研究强调了选择性肌苷加油增强CART治疗实体瘤疗效的潜力.
    Chimeric antigen receptor (CAR) T cell therapy is hindered in solid tumor treatment due to the immunosuppressive tumor microenvironment and suboptimal T cell persistence. Current strategies do not address nutrient competition in the microenvironment. Hence, we present a metabolic refueling approach using inosine as an alternative fuel. CAR T cells were engineered to express membrane-bound CD26 and cytoplasmic adenosine deaminase 1 (ADA1), converting adenosine to inosine. Autocrine secretion of ADA1 upon CD3/CD26 stimulation activates CAR T cells, improving migration and resistance to transforming growth factor β1 suppression. Fusion of ADA1 with anti-CD3 scFv further boosts inosine production and minimizes tumor cell feeding. In mouse models of hepatocellular carcinoma and non-small cell lung cancer, metabolically refueled CAR T cells exhibit superior tumor reduction compared to unmodified CAR T cells. Overall, our study highlights the potential of selective inosine refueling to enhance CAR T therapy efficacy against solid tumors.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号