MT: Regular Issue

  • 文章类型: Journal Article
    嵌合抗原受体(CAR)T细胞对实体瘤的成功有限。这里,我们使用溶瘤泡沫病毒(oFV)与抗CD19CART细胞联合在肿瘤上展示模型CAR靶抗原(CD19).我们产生了oFV-Δbel2和oFV-bel2载体以测试病毒/CD19传播的效率和稳定性。虽然这两种病毒在体外具有相同的CAR-T杀伤能力,OFV-Δbel2病毒获得了G到A突变,而oFV-bel2病毒有基因组缺失。在体内皮下肿瘤模型中,CART细胞导致OFV特异性生物发光的显著降低,确认OFV感染的肿瘤细胞的清除。然而,最有效的治疗是在缺乏CAR-T细胞的情况下使用高剂量OFV,表明OFV的CART清除是有害的。此外,在逃避CART细胞治疗的肿瘤中,复发病毒在OFV-CD19转基因中含有缺失,允许病毒逃避CART消除。因此,OFV代表一种缓慢阴燃的溶瘤病毒,其通过肿瘤的慢性扩散产生了抗肿瘤治疗,CART疗法废除了。这些结果表明,该溶瘤平台的进一步发展,有了额外的免疫治疗武器,可能允许慢性溶瘤的有效组合。
    Chimeric antigen receptor (CAR) T cells have had limited success against solid tumors. Here, we used an oncolytic foamy virus (oFV) to display a model CAR target antigen (CD19) on tumors in combination with anti-CD19 CAR T cells. We generated oFV-Δbel2 and oFV-bel2 vectors to test the efficiency and stability of viral/CD19 spread. While both viruses conferred equal CAR T killing in vitro, the oFV-Δbel2 virus acquired G-to-A mutations, whereas oFV-bel2 virus had genome deletions. In subcutaneous tumor models in vivo, CAR T cells led to a significant decrease in oFV-specific bioluminescence, confirming clearance of oFV-infected tumor cells. However, the most effective therapy was with high-dose oFV in the absence of CAR T cells, indicating that CAR T clearance of oFV was detrimental. Moreover, in tumors that escaped CAR T cell treatment, resurgent virus contained deletions within the oFV-CD19 transgene, allowing the virus to escape CAR T elimination. Therefore, oFV represents a slow smoldering type of oncolytic virus, whose chronic spread through tumors generates anti-tumor therapy, which is abolished by CAR T therapy. These results suggest that further development of this oncolytic platform, with additional immunotherapeutic arming, may allow for an effective combination of chronic oncolysis.
    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
    靶向T细胞上的免疫检查点受体是常见的癌症治疗策略。经常,这是通过靶向抑制性共受体配体的抗体实现的。阻断免疫检查点PD-1与其配体PD-L1和PD-L2的结合防止下游信号传导并增强抗肿瘤T细胞应答。这种方法改善了癌症患者的预后。然而,只有三分之一的患者对这些治疗有反应。为了更好地了解抗PD-1抗体的机制,我们探索了PD-1在免疫突触中的位置。令人惊讶的是,我们发现抗PD-1抗体,除了阻断PD-1和它的配体之间的相互作用,也从突触中去除PD-1。我们证明了通过抗PD-1抗体从突触中去除PD-1与T细胞活化程度之间的相关性。有趣的是,短版本的抗PD-1抗体,F(ab')2未能从突触中移除PD-1并激活T细胞。使用同系肿瘤模型,我们显示抗PD-1抗体的抗肿瘤效果优于较短版本的相同抗体.我们的数据表明,抗PD-1抗体通过从突触中去除PD-1激活T细胞,改变PD-1或其他免疫受体在免疫突触中的位置可以作为替代方案,治疗癌症的有效方法。
    Targeting immune checkpoint receptors on T cells is a common cancer treatment strategy. Frequently, this is accomplished through antibodies targeting the ligand of inhibitory co-receptors. Blocking the immune checkpoint PD-1 binding to its ligands PD-L1 and PD-L2 prevents downstream signaling and enhances anti-tumor T cell responses. This approach improves cancer patients\' outcomes. However, only one-third of the patients respond to these treatments. To better understand the mechanism of anti-PD-1 antibodies, we explored the location of PD-1 within the immune synapse. Surprisingly, we discovered that anti-PD-1 antibodies, besides blocking the interaction between PD-1 and its ligands, also removed PD-1 from the synapse. We demonstrated a correlation between removing PD-1 from the synapse by anti-PD-1 antibodies and the extent of T cell activation. Interestingly, a short version of the anti-PD-1 antibody, F(ab\')2, failed to remove PD-1 from the synapse and activate T cells. Using the syngeneic tumor model, we showed a superior anti-tumor effect of the anti-PD-1 antibody over the shorter version of the same antibody. Our data indicate that anti-PD-1 antibodies activate T cells by removing PD-1 from the synapse, and changing the location of PD-1 or other immune receptors within the immune synapse could serve as an alternative, efficient approach to treat cancer.
    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
    迄今为止,近四分之一的结直肠癌(CRC)患者发生肝转移(CRCLM),其侵袭性可以与确定的组织病理学生长模式(HGP)相关。从CRCLM内的三个主要HGP来看,替代的HGP显得特别激进,以增强的肿瘤细胞运动和血管共选择为特征。这里,我们研究了钙和整合素结合蛋白1(CIB1)的表达之间的相关性,广泛表达的基因涉及各种细胞过程,包括迁移和粘附,原发性CRC患者的无病生存期(DFS)和总生存期(OS)。此外,我们探讨了CRCLM不同HGPs与CIB1表达的相关性。蛋白质组学分析用于评估697名原发性CRC患者队列中的CIB1表达。此外,单细胞和空间RNA测序数据集,使用公开的批量测序数据评估CRCLM中CIB1的表达。通过福尔马林固定的石蜡包埋的免疫组织化学染色进一步验证了计算机数据。我们观察到高CIB1表达与较差的DFS和OS独立相关,不管国际联盟对抗癌症阶段,性别,或年龄。此外,侵袭性替代CRCLMHGP与高CIB1表达显著相关。我们的发现表明CIB1水平与CRC的临床侵袭性之间存在相关性。此外,CIB1可能是对HGPCRCLM进行分层的新标记。
    To date, nearly one-quarter of colorectal cancer (CRC) patients develop liver metastases (CRCLM), and its aggressiveness can be correlated to defined histopathological growth patterns (HGP). From the three main HGPs within CRCLM, the replacement HGP emerges as particularly aggressive, characterized by heightened tumor cell motility and vessel co-option. Here, we investigated the correlation between the expression of calcium- and integrin-binding protein 1 (CIB1), a ubiquitously expressed gene involved in various cellular processes including migration and adhesion, and disease-free (DFS) and overall survival (OS) in primary CRC patients. Additionally, we explored the correlation between CIB1 expression and different HGPs of CRCLM. Proteomic analysis was used to evaluate CIB1 expression in a cohort of 697 primary CRC patients. Additionally, single-cell and spatial RNA-sequencing datasets, along with publicly available bulk sequencing data were used to evaluate CIB1 expression in CRCLM. In silico data were further validated by formalin-fixed paraffin-embedded immunohistochemical stainings. We observed that high CIB1 expression is independently associated with worse DFS and OS, regardless of Union Internationale Contre le Cancer stage, gender, or age. Furthermore, the aggressive replacement CRCLM HGP is significantly associated with high CIB1 expression. Our findings show a correlation between CIB1 levels and the clinical aggressiveness of CRC. Moreover, CIB1 may be a novel marker to stratify HGP CRCLM.
    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
    CD19嵌合抗原受体T(CD19CAR-T)细胞在复发性/难治性B细胞恶性肿瘤中取得了有希望的结果。然而,复发的发生是由于CAR-T细胞持久性的丧失。我们在CAR-T细胞中开发了双重T/B细胞共刺激分子(CD28和CD40),以增强强烈的杀肿瘤活性和持久性。CD19.28.40zCAR-T细胞促进pNF-κB和pRelB下游信号传导,同时在抗原暴露时减少NFAT信号传导。CD19.28.40zCAR-T细胞表现出更大的增殖,在长期共培养试验中转化为有效的抗肿瘤细胞毒性。每周重复的抗原刺激揭示了连续的CAR-T细胞扩增,同时保留了中枢记忆T细胞亚群和较低的耗尽表型表达。将CD19.28.40zCAR-T细胞反应的内在基因与常规CAR进行了比较,并证明了与T细胞增殖和记忆相关的上调基因以及与凋亡相关的下调基因。疲惫,和糖酵解途径。T细胞干细胞基因的富集,特别是出售,IL-7r,观察到TCF7和KLF2。在B细胞淋巴细胞白血病和B细胞非霍奇金淋巴瘤异种移植模型中都表现出有效和持续的体内抗肿瘤细胞毒性,同时表现出持续的T细胞记忆特征。进一步验证了CD37.28.40zCAR-T细胞针对CD37+肿瘤细胞的活性的功能增强。双T/B细胞信号分子的修饰显著地最大化了CAR-T细胞疗法的功效。
    CD19 chimeric antigen receptor T (CD19CAR-T) cells have achieved promising outcomes in relapsed/refractory B cell malignancies. However, recurrences occur due to the loss of CAR-T cell persistence. We developed dual T/B cell co-stimulatory molecules (CD28 and CD40) in CAR-T cells to enhance intense tumoricidal activity and persistence. CD19.28.40z CAR-T cells promoted pNF-κB and pRelB downstream signaling while diminishing NFAT signaling upon antigen exposure. CD19.28.40z CAR-T cells demonstrated greater proliferation, which translated into effective anti-tumor cytotoxicity in long-term co-culture assay. Repetitive weekly antigen stimulation unveiled continuous CAR-T cell expansion while preserving central memory T cell subset and lower expression of exhaustion phenotypes. The intrinsic genes underlying CD19.28.40z CAR-T cell responses were compared with conventional CARs and demonstrated the up-regulated genes associated with T cell proliferation and memory as well as down-regulated genes related to apoptosis, exhaustion, and glycolysis pathway. Enrichment of genes toward T cell stemness, particularly SELL, IL-7r, TCF7, and KLF2, was observed. Effective and continuing anti-tumor cytotoxicity in vivo was exhibited in both B cell lymphoblastic leukemia and B cell non-Hodgkin lymphoma xenograft models while demonstrating persistent T cell memory signatures. The functional enhancement of CD37.28.40z CAR-T cell activities against CD37+ tumor cells was further validated. The modification of dual T/B cell signaling molecules remarkably maximized the efficacy of CAR-T 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
    肿瘤归巢神经干细胞(NSC)疗法正在成为侵袭性脑癌的有希望的治疗方法。尽管他们取得了成功,开发肿瘤归巢NSC治疗疗法,保持持久的肿瘤抑制仍然是一个挑战。在这里,我们报道了一种协同联合治疗方案,其中新型小分子TR-107增强了NSC-肿瘤坏死因子相关凋亡诱导配体(TRAIL)治疗(hiNeuroS-TRAIL)在体外无法治愈的脑癌胶质母细胞瘤(GBM)模型中的作用.我们报告说,hiNeuroS-TRAIL和TR-107的组合协同上调caspase标志物,并通过显着下调与TRAIL抗性LN229细胞系中的化学抗性和放射抗性相关的抑制途径来恢复对内在凋亡途径的敏感性。这种组合还显示出强大的肿瘤抑制和增强小鼠的存活率,这些发现阐明了一种新的组合方案,并表明这些临床相关药物的组合可能代表一种新的治疗选择,对GBM患者具有更高的疗效。
    Tumor-homing neural stem cell (NSC) therapy is emerging as a promising treatment for aggressive cancers of the brain. Despite their success, developing tumor-homing NSC therapy therapies that maintain durable tumor suppression remains a challenge. Herein, we report a synergistic combination regimen where the novel small molecule TR-107 augments NSC-tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) therapy (hiNeuroS-TRAIL) in models of the incurable brain cancer glioblastoma (GBM) in vitro. We report that the combination of hiNeuroS-TRAIL and TR-107 synergistically upregulated caspase markers and restored sensitivity to the intrinsic apoptotic pathway by significantly downregulating inhibitory pathways associated with chemoresistance and radioresistance in the TRAIL-resistant LN229 cell line. This combination also showed robust tumor suppression and enhanced survival of mice bearing human xenografts of both solid and invasive GBMs. These findings elucidate a novel combination regimen and suggest that the combination of these clinically relevant agents may represent a new therapeutic option with increased efficacy for patients with GBM.
    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
    通过小分子对细胞-细胞相互作用的邻近诱导代表了基础和翻译科学中的新兴领域。这些小分子的共价锚定代表了一种有用的化学策略来加强接近度;然而,它在很大程度上仍未被探索驱动细胞-细胞相互作用。在免疫治疗应用中,双功能小分子是诱导免疫效应细胞如T细胞和肿瘤细胞之间接近以诱导杀肿瘤功能的有吸引力的工具。我们描述了一种由亲电双功能小分子和配对的合成抗原受体(SAR)组成的双组分系统,可引起T细胞活化。分子,被称为共价免疫招募者(CIR),被设计为亲和标记并共价接合SAR。我们评估了CIRs在指导使用三种生物学上不同类型的SAR工程的人T细胞的抗肿瘤功能方面的实用性。不考虑亲电子化学,肿瘤靶向部分,或SAR设计,CIRs优于等效的非共价双功能衔接子,在最大化功能方面确立了共价的关键作用。我们确定共价连接以依赖于每个SAR生物学和信号传导阈值的方式强制早期T细胞活化事件。这些结果提供了优化通用SAR-T细胞功能的平台,并更广泛地揭示了共价衔接子如何调节细胞-细胞邻近诱导的新见解。
    Proximity-induction of cell-cell interactions via small molecules represents an emerging field in basic and translational sciences. Covalent anchoring of these small molecules represents a useful chemical strategy to enforce proximity; however, it remains largely unexplored for driving cell-cell interactions. In immunotherapeutic applications, bifunctional small molecules are attractive tools for inducing proximity between immune effector cells like T cells and tumor cells to induce tumoricidal function. We describe a two-component system composed of electrophilic bifunctional small molecules and paired synthetic antigen receptors (SARs) that elicit T cell activation. The molecules, termed covalent immune recruiters (CIRs), were designed to affinity label and covalently engage SARs. We evaluated the utility of CIRs to direct anti-tumor function of human T cells engineered with three biologically distinct classes of SAR. Irrespective of the electrophilic chemistry, tumor-targeting moiety, or SAR design, CIRs outperformed equivalent non-covalent bifunctional adapters, establishing a key role for covalency in maximizing functionality. We determined that covalent linkage enforced early T cell activation events in a manner that was dependent upon each SARs biology and signaling threshold. These results provide a platform to optimize universal SAR-T cell functionality and more broadly reveal new insights into how covalent adapters modulate cell-cell proximity-induction.
    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
    肉瘤是一种罕见且高度多样化的间充质起源的恶性肿瘤。虽然肉瘤通常被认为对免疫疗法有抗性,最近的研究表明,对检查点抑制剂(CPIs)的临床应答存在亚型特异性差异,这与肉瘤亚型中存在的不同免疫表型相关.溶瘤病毒(OVs)被设计为选择性感染和杀死肿瘤细胞,并诱导肿瘤内免疫浸润,增强免疫原性,从而使肿瘤对免疫疗法敏感。在此,我们回顾了评估肉瘤OVs的累积临床数据。少数肉瘤患者被纳入早期OV试验,作为较大实体瘤队列的一部分,证明安全性,但由于患者人数少和缺乏组织学分组,对生物学效应的了解有限。最近的几项研究已经调查了talimogenelaherparepvec(T-VEC),一种经批准的溶瘤单纯疱疹病毒(HSV-1),在肉瘤患者队列的联合治疗方案中。这些研究表明,在严重预处理和免疫疗法抵抗的患者中,有希望的反应与肿瘤内免疫浸润增加有关。随着新的更有效的OVs进入临床领域,在亚型特异性队列中进行前瞻性评估,并进行相关研究以确定应答的生物标志物,这对于推进这种有前景的肉瘤治疗方法至关重要.
    Sarcomas are a rare and highly diverse group of malignancies of mesenchymal origin. While sarcomas are generally considered resistant to immunotherapy, recent studies indicate subtype-specific differences in clinical response to checkpoint inhibitors (CPIs) that are associated with distinct immune phenotypes present in sarcoma subtypes. Oncolytic viruses (OVs) are designed to selectively infect and kill tumor cells and induce intratumoral immune infiltration, enhancing immunogenicity and thereby sensitizing tumors to immunotherapy. Herein we review the accumulated clinical data evaluating OVs in sarcoma. Small numbers of patients with sarcoma were enrolled in early-stage OV trials as part of larger solid tumor cohorts demonstrating safety but providing limited insight into the biological effects due to the low patient numbers and lack of histologic grouping. Several recent studies have investigated talimogene laherparepvec (T-VEC), an approved oncolytic herpes simplex virus (HSV-1), in combination therapy regimens in sarcoma patient cohorts. These studies have shown promising responses in heavily pre-treated and immunotherapy-resistant patients associated with increased intratumoral immune infiltration. As new and more potent OVs enter the clinical arena, prospective evaluation in subtype-specific cohorts with correlative studies to define biomarkers of response will be critical to advancing this promising approach for sarcoma 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
    CD4+T辅助抗原是癌症疫苗的重要组成部分,但这些MHCII类限制性抗原来源的相关性仍未得到充分研究。为了比较肿瘤特异性辅助抗原与肿瘤无关辅助抗原的有效性,我们为小鼠MC-38结肠癌设计了三种DNA疫苗,单独编码CD8+T细胞新抗原(noHELP)或与“通用”辅助抗原(uniHELP)或辅助新抗原(neoHELP)联合编码。两种类型的帮助疫苗都增加了疫苗诱导的CD8+T细胞的频率,特别是uniHELP增加了KLRG1+和PD-1低效应细胞的比例。然而,当小鼠随后注射MC-38细胞时,只有neoHELP疫苗接种的肿瘤控制效果明显优于noHELP。与uniHELP相比,neoHELP诱导的肿瘤控制依赖于CD4+T细胞的存在,而两种疫苗都依赖于CD8+T细胞。与此相符,含有CD4+或CD8+T细胞新抗原的野生型对应物的neoHELP变体显示出降低的肿瘤控制。这些数据表明最佳的个性化癌症疫苗应包括MHCII类限制性新抗原以引发肿瘤特异性CD4+T细胞帮助。
    CD4+ T helper antigens are essential components of cancer vaccines, but the relevance of the source of these MHC class II-restricted antigens remains underexplored. To compare the effectiveness of tumor-specific versus tumor-unrelated helper antigens, we designed three DNA vaccines for the murine MC-38 colon carcinoma, encoding CD8+ T cell neoantigens alone (noHELP) or in combination with either \"universal\" helper antigens (uniHELP) or helper neoantigens (neoHELP). Both types of helped vaccines increased the frequency of vaccine-induced CD8+ T cells, and particularly uniHELP increased the fraction of KLRG1+ and PD-1low effector cells. However, when mice were subsequently injected with MC-38 cells, only neoHELP vaccination resulted in significantly better tumor control than noHELP. In contrast to uniHELP, neoHELP-induced tumor control was dependent on the presence of CD4+ T cells, while both vaccines relied on CD8+ T cells. In line with this, neoHELP variants containing wild-type counterparts of the CD4+ or CD8+ T cell neoantigens displayed reduced tumor control. These data indicate that optimal personalized cancer vaccines should include MHC class II-restricted neoantigens to elicit tumor-specific CD4+ T cell help.
    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
    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
    双特异性T细胞衔接剂是一类有前途的用于癌症治疗的治疗性蛋白质。它们的效力和小尺寸通常伴随着全身毒性和短半衰期,使静脉给药变得繁琐。这些限制可以通过肿瘤特异性原位表达来克服,允许高局部积累,同时降低全身浓度。然而,编码病毒或非病毒载体中的T细胞衔接者并原位表达它们消除了在重组蛋白生产期间进行的所有形式的质量控制。因此,设计具有最小域错误配对的构造至关重要,和增加的同质性的治疗产品。这里,我们报道了一种专门为载体介导的免疫治疗而设计的T细胞接合器结构.它基于设计的锚蛋白重复蛋白(DARPin)与CD3靶向单链抗体片段的融合,称为DATE(DARPin融合T细胞接合器)。DATE诱导强效T细胞介导的HER2+癌细胞杀伤,作为重组产生的治疗性蛋白和作为来自HER2+重靶向的高容量腺病毒载体(HC-AdV)的原位表达的有效载荷。我们报告了显著的肿瘤缓解,日期累积,和T细胞浸润通过在体内由HER2+重新靶向的HC-AdV介导的原位表达。我们的结果支持DATEs作为载体介导的免疫治疗的有效载荷的进一步研究和发展。
    Bispecific T cell engagers are a promising class of therapeutic proteins for cancer therapy. Their potency and small size often come with systemic toxicity and short half-life, making intravenous administration cumbersome. These limitations can be overcome by tumor-specific in situ expression, allowing high local accumulation while reducing systemic concentrations. However, encoding T cell engagers in viral or non-viral vectors and expressing them in situ ablates all forms of quality control performed during recombinant protein production. It is therefore vital to design constructs that feature minimal domain mispairing, and increased homogeneity of the therapeutic product. Here, we report a T cell engager architecture specifically designed for vector-mediated immunotherapy. It is based on a fusion of a designed ankyrin repeat protein (DARPin) to a CD3-targeting single-chain antibody fragment, termed DATE (DARPin-fused T cell Engager). The DATE induces potent T cell-mediated killing of HER2+ cancer cells, both as recombinantly produced therapeutic protein and as in situ expressed payload from a HER2+-retargeted high-capacity adenoviral vector (HC-AdV). We report remarkable tumor remission, DATE accumulation, and T cell infiltration through in situ expression mediated by a HER2+-retargeted HC-AdV in vivo. Our results support further investigations and developments of DATEs as payloads for vector-mediated immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号