Receptor Tyrosine Kinase-like Orphan Receptors

  • 文章类型: Journal Article
    本研究旨在探讨受体酪氨酸激酶样孤儿受体2(ROR2)在三阴性乳腺癌(TNBC)中的作用。
    通过免疫组织化学染色和PCR分析原发性TNBC和转移性TNBC组织中的ROR2表达。通过PCR和Western印迹分析检测TNBC细胞系中的ROR2表达。移民,检测ROR2过表达或敲低的TNBC细胞的侵袭和化学敏感性。
    ROR2在转移性TNBC组织中高表达。ROR2击倒抑制了迁移,TNBC细胞的侵袭和化学抗性。ROR2过表达促进MDA-MB-435细胞的迁移,入侵,和化学抗性。此外,HC1599和MDA-MB-435阿霉素耐药细胞中的ROR2敲低增强了对阿霉素的化学敏感性。ROR2可以激活TNBC细胞中的PI3K/AKT/mTOR信号。
    ROR2上调并通过激活PI3K/AKT/mTOR信号促进TNBC的转移表型。
    UNASSIGNED: This study aimed to investigate the role of receptor tyrosine kinase-like orphan receptor 2 (ROR2) in triple-negative breast cancer (TNBC).
    UNASSIGNED: ROR2 expression in primary TNBC and metastatic TNBC tissues was analyzed by immunohistochemical staining and PCR. ROR2 expression in TNBC cell lines was detected by PCR and Western blot analysis. The migration, invasion and chemosensitivity of TNBC cells with overexpression or knockdown of ROR2 were examined.
    UNASSIGNED: ROR2 expression was high in metastatic TNBC tissues. ROR2 knockdown suppressed the migration, invasion and chemoresistance of TNBC cells. ROR2 overexpression in MDA-MB-435 cells promoted the migration, invasion, and chemoresistance. Moreover, ROR2 knockdown in HC1599 and MDA-MB-435 adriamycin-resistant cells enhanced chemosensitivity to adriamycin. ROR2 could activate PI3K/AKT/mTOR signaling in TNBC cells.
    UNASSIGNED: ROR2 is upregulated and promotes metastatic phenotypes of TNBC by activating PI3K/AKT/mTOR signaling.
    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
    细胞极性机制允许形成具有独特蛋白质组成的专门膜结构域,信号属性,和功能特征。通过分析钾通道和属于肌营养不良蛋白相关蛋白复合物的蛋白质的定位,我们揭示了在C.elegans肌肉细胞表面存在不同的平面极化膜区室。我们发现,肌肉极性是由涉及配体EGL-20/Wnt的非规范Wnt信号级联控制的,受体CAM-1/Ror,和细胞内效应物DSH-1/Dishevelled。有趣的是,此过程不需要经典的平面细胞极性蛋白。使用时间分辨的蛋白质降解,我们证明了-虽然它在胚胎发生结束时基本上已经到位-肌肉极性是一种动态状态,需要在整个胚胎后生命中持续存在DSH-1。我们的结果揭示了C.elegans肌肉膜的意外复杂性,并建立了一个可遗传处理的模型系统来研究体内细胞极性和膜区室化。
    Cell polarity mechanisms allow the formation of specialized membrane domains with unique protein compositions, signalling properties, and functional characteristics. By analyzing the localization of potassium channels and proteins belonging to the dystrophin-associated protein complex, we reveal the existence of distinct planar-polarized membrane compartments at the surface of C. elegans muscle cells. We find that muscle polarity is controlled by a non-canonical Wnt signalling cascade involving the ligand EGL-20/Wnt, the receptor CAM-1/Ror, and the intracellular effector DSH-1/Dishevelled. Interestingly, classical planar cell polarity proteins are not required for this process. Using time-resolved protein degradation, we demonstrate that -while it is essentially in place by the end of embryogenesis- muscle polarity is a dynamic state, requiring continued presence of DSH-1 throughout post-embryonic life. Our results reveal the unsuspected complexity of the C. elegans muscle membrane and establish a genetically tractable model system to study cellular polarity and membrane compartmentalization 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
    胰腺导管腺癌(PDAC)是最致命的癌症之一,腹膜播散是预后不良的主要原因之一。外泌体已经成为胃肠道癌症的有希望的生物标志物,可以在各种体液中发现,也在腹膜液(PF)。这是一个独特的样品,因为它接近胃肠道恶性肿瘤。受体酪氨酸激酶样孤儿受体1(ROR1)已被确定为人类癌症的潜在生物标志物,并代表了免疫治疗方法的有希望的靶标。这可以考虑为未来的治疗策略。在这里,我们前瞻性地分析了一方面是局部PDAC患者(PER-)中PF中的外泌体表面蛋白ROR1(exo-ROR1),另一方面是腹膜播散性肿瘤分期(PER),然后是exo-ROR1与临床病理参数的相关性。
    从PF和非癌(NC)的血浆样品中分离外泌体(n=15),慢性胰腺炎(CP)(n=4),局部PDAC(PER-)(n=18)和腹膜播散性PDAC(PER+)(n=9)患者,通过FACS分析检测到表面蛋白ROR1.此外,分析了PF中可溶性ROR1。使用蛋白质印迹(WB)研究组织中的ROR1表达,qPCR,免疫组织化学(IHC)。通过纳米跟踪分析(NTA)证明了外泌体隔离,WB,透射电子显微镜(TEM),和BCA蛋白测定。结果与临床资料相关,并进行生存分析。
    PDAC(PER)患者在PF中具有最高的exo-ROR1值,可以与NC区分开(p<0.0001),PDAC(PER-)(p<0.0001),和CP(p=0.0112)。PDAC(PER-)可以与NC(p=0.0003)区分开。在等离子体中,exo-ROR1不能区分这些基团。虽然在胰腺外分泌组织中没有ROR1的表达,PDAC和腹膜转移显示ROR1表达。PF中的高exo-ROR1表达与较低的总生存率相关(p=0.0482)。
    在PF中使用exo-ROR1,我们发现了一种有希望的诊断和预后生物标志物,可能区分NC,PDAC(PER-)和PDAC(PER+),并可能阐明PDAC未来的诊断和治疗概念。
    UNASSIGNED: Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest forms of cancer and peritoneal dissemination is one major cause for this poor prognosis. Exosomes have emerged as promising biomarkers for gastrointestinal cancers and can be found in all kinds of bodily fluids, also in peritoneal fluid (PF). This is a unique sample due to its closeness to gastrointestinal malignancies. The receptor tyrosine kinase-like orphan receptor 1 (ROR1) has been identified as a potential biomarker in human cancers and represents a promising target for an immunotherapy approach, which could be considered for future treatment strategies. Here we prospectively analyzed the exosomal surface protein ROR1 (exo-ROR1) in PF in localized PDAC patients (PER-) on the one hand and peritoneal disseminated tumor stages (PER+) on the other hand followed by the correlation of exo-ROR1 with clinical-pathological parameters.
    UNASSIGNED: Exosomes were isolated from PF and plasma samples of non-cancerous (NC) (n = 15), chronic pancreatitis (CP) (n = 4), localized PDAC (PER-) (n = 18) and peritoneal disseminated PDAC (PER+) (n = 9) patients and the surface protein ROR1 was detected via FACS analysis. Additionally, soluble ROR1 in PF was analyzed. ROR1 expression in tissue was investigated using western blots (WB), qPCR, and immunohistochemistry (IHC). Exosome isolation was proven by Nano Tracking Analysis (NTA), WB, Transmission electron microscopy (TEM), and BCA protein assay. The results were correlated with clinical data and survival analysis was performed.
    UNASSIGNED: PDAC (PER+) patients have the highest exo-ROR1 values in PF and can be discriminated from NC (p <0.0001), PDAC (PER-) (p <0.0001), and CP (p = 0.0112). PDAC (PER-) can be discriminated from NC (p = 0.0003). In plasma, exo-ROR1 is not able to distinguish between the groups. While there is no expression of ROR1 in the exocrine pancreatic tissue, PDAC and peritoneal metastasis show expression of ROR1. High exo-ROR1 expression in PF is associated with lower overall survival (p = 0.0482).
    UNASSIGNED: With exo-ROR1 in PF we found a promising diagnostic and prognostic biomarker possibly discriminating between NC, PDAC (PER-) and PDAC (PER+) and might shed light on future diagnostic and therapeutic concepts in PDAC.
    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
    受体酪氨酸激酶ROR2介导非规范的WNT5A信号,以协调组织形态发生过程,通路的功能障碍导致Robinow综合征,短指B和转移性疾病。ROR2功能所需的域和机制,然而,仍然不清楚。我们解决了ROR2的细胞外富含半胱氨酸(CRD)和Kringle(Kr)结构域的晶体结构,与其他CRD不同,ROR2CRD缺乏结合脂质/脂质修饰蛋白的特征疏水口袋,例如WNTs,提出了一种新的配体接收机制。功能上,我们显示了ROR2CRD,但不是其他领域,是必需的,并且最低限度地足以促进WNT5A信令,CRD和邻近Kr中的Robinow突变会损害ROR2的分泌和功能。此外,使用针对WNT受体的卷曲(FZ)家族的功能激活和干扰抗体,我们证明了FZ参与WNT5A-ROR信号传导。因此,ROR2通过其CRD起作用以增强包括FZ的受体超复合物的功能以转导WNT5A信号。
    The receptor tyrosine kinase ROR2 mediates noncanonical WNT5A signaling to orchestrate tissue morphogenetic processes, and dysfunction of the pathway causes Robinow syndrome, brachydactyly B, and metastatic diseases. The domain(s) and mechanisms required for ROR2 function, however, remain unclear. We solved the crystal structure of the extracellular cysteine-rich (CRD) and Kringle (Kr) domains of ROR2 and found that, unlike other CRDs, the ROR2 CRD lacks the signature hydrophobic pocket that binds lipids/lipid-modified proteins, such as WNTs, suggesting a novel mechanism of ligand reception. Functionally, we showed that the ROR2 CRD, but not other domains, is required and minimally sufficient to promote WNT5A signaling, and Robinow mutations in the CRD and the adjacent Kr impair ROR2 secretion and function. Moreover, using function-activating and -perturbing antibodies against the Frizzled (FZ) family of WNT receptors, we demonstrate the involvement of FZ in WNT5A-ROR signaling. Thus, ROR2 acts via its CRD to potentiate the function of a receptor super-complex that includes FZ to transduce WNT5A signals.
    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
    髓母细胞瘤(MB),一种常见的小儿中枢神经系统肿瘤,受影响肿瘤起始和进展的微小RNA(miRNA)的影响。然而,miRNAs在MB肿瘤发生中的特异性参与尚不清楚.使用单细胞RNA测序,我们确定了正常人胎儿小脑中ROR2的表达。随后的分析,包括免疫荧光,实时定量PCR(qRT-PCR),和蛋白质印迹,评估MB组织和细胞系中的ROR2表达。我们通过双荧光素酶报告基因研究了miR-124-3p和miR-194-5p及其在ROR2表达中的调节作用。qRT-PCR,和蛋白质印迹分析。通过探索miR-124-3p的影响的功能测定获得了机制见解,miR-194-5p,和ROR2对MB体外和体内生长的影响。我们在MB组织和细胞系中观察到miR-124-3p和miR-194-5p表达显着降低,ROR2表达升高。在WNT和SHH亚组的MB患者中,高ROR2表达与总生存期呈负相关。功能上,过表达miR-124-3p和miR-194-5p并抑制ROR2抑制了体外恶性转化和体内致瘤性。机械上,miR-124-3p和miR-194-5p协同调控ROR2/PI3K/Akt通路,影响MB进展。我们的发现表明miR-124-3p和miR-194-5p作为肿瘤抑制因子,通过ROR2/PI3K/Akt轴抑制MB进展,提示MB患者的关键机制和治疗目标。
    Medulloblastoma (MB), a prevalent pediatric central nervous system tumor, is influenced by microRNAs (miRNAs) that impact tumor initiation and progression. However, the specific involvement of miRNAs in MB tumorigenesis remains unclear. Using single-cell RNA sequencing, we identified ROR2 expression in normal human fetal cerebellum. Subsequent analyses, including immunofluorescence, quantitative real-time PCR (qRT-PCR), and Western blot, assessed ROR2 expression in MB tissues and cell lines. We investigated miR-124-3p and miR-194-5p and their regulatory role in ROR2 expression through the dual-luciferase reporter, qRT-PCR, and western blot assays. Mechanistic insights were gained through functional assays exploring the impact of miR-124-3p, miR-194-5p, and ROR2 on MB growth in vitro and in vivo. We observed significantly reduced miR-124-3p and miR-194-5p expression and elevated ROR2 expression in MB tissues and cell lines. High ROR2 expression inversely correlated with overall survival in WNT and SHH subgroups of MB patients. Functionally, overexpressing miR-124-3p and miR-194-5p and inhibiting ROR2 suppressed in vitro malignant transformation and in vivo tumorigenicity. Mechanistically, miR-124-3p and miR-194-5p synergistically regulated the ROR2/PI3K/Akt pathway, influencing MB progression. Our findings indicate that miR-124-3p and miR-194-5p function as tumor suppressors, inhibiting MB progression via the ROR2/PI3K/Akt axis, suggesting a key mechanism and therapeutic targets for MB patients.
    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:嵌合抗原受体(CAR)T细胞治疗靶受体酪氨酸激酶样孤儿受体1(ROR1)在血液肿瘤和实体瘤中广泛表达,然而,具有单链可变片段(scFv)-R12靶向域的临床特征的ROR1-CART细胞未能诱导持久的缓解,部分是由于免疫抑制肿瘤微环境(TME)。在这里,我们描述了一种改进的ROR1-CAR的开发,全人scFv9靶向域,并增强了TGFβRIIDN装甲,可抵抗主要的TME因素,转化生长因子β(TGFβ)。
    方法:通过慢病毒转导富集的CD4+和CD8+T细胞产生CART细胞,并且在体外和体内将新型的基于scFv9的ROR1-CAR-1与临床表征的基于ROR1-R12-scFv的CAR-2进行了比较。
    结果:CAR-1T细胞表现出比CAR-2更高的CAR表面密度,并产生更多的干扰素(IFN)-γ肿瘤坏死因子(TNF)-α和白细胞介素(IL)-2响应于血液学(Jeko-1,RPMI-8226)和实体(OVCAR-3,Capan-2,NCI-H226)体外肿瘤细胞系。在体内,CAR-1和CAR-2均清除了血液系统Jeko-1淋巴瘤异种移植物,然而,只有CAR-1完全拒绝卵巢实体OVCAR-3肿瘤,与CD8+和CD4+CART细胞的更大扩增相一致,并富集中枢和效应记忆表型。当配备TGFβ防护装甲TGFβRIIDN时,CAR-1T细胞抵抗TGFβ介导的pSmad2/3磷酸化,与单独的CAR-1相比。当与ROR-1+AsPC-1胰腺癌细胞系在TGFβ1的存在下共培养时,装甲CAR-1表现出改善的杀伤功能恢复,IFN-γ,TNF-α和IL-2分泌。与单独的CAR-1相比,在过表达TGFβ1的小鼠AsPC-1胰腺肿瘤异种移植物中,装甲的CAR-1,实现了完整的肿瘤缓解,加速了CAR+T细胞的扩增,循环活性TGFβ1减少,无明显毒性或体重减轻。出乎意料的是,在无TGFβ过表达的AsPC-1异种移植物中,ROR-1-CART细胞与ROR-1阳性肿瘤细胞相互作用特异性诱导TGFβ1的产生,TGFβRIIDN装甲加速了肿瘤清除。
    结论:新型的完全人TGFβRIIDN装甲的ROR1-CAR-1T细胞对ROR1阳性肿瘤具有高度的效力,并耐受TGFβ在固体TME中的抑制作用。此外,TGFβ1诱导代表了一种新的,固体TME中的CAR诱导检查点,这可以通过在T细胞上共表达TGβRIIDN装甲来规避。
    BACKGROUND: Chimeric antigen receptor (CAR) T-cell therapy target receptor tyrosine kinase-like orphan receptor 1 (ROR1) is broadly expressed in hematologic and solid tumors, however clinically-characterized ROR1-CAR T cells with single chain variable fragment (scFv)-R12 targeting domain failed to induce durable remissions, in part due to the immunosuppressive tumor microenvironment (TME). Herein, we describe the development of an improved ROR1-CAR with a novel, fully human scFv9 targeting domain, and augmented with TGFβRIIDN armor protective against a major TME factor, transforming growth factor beta (TGFβ).
    METHODS: CAR T cells were generated by lentiviral transduction of enriched CD4+ and CD8+ T cells, and the novel scFv9-based ROR1-CAR-1 was compared with the clinically-characterized ROR1-R12-scFv-based CAR-2 in vitro and in vivo.
    RESULTS: CAR-1 T cells exhibited greater CAR surface density than CAR-2 when normalized for %CAR+, and produced more interferon (IFN)-γ tumor necrosis factor (TNF)-α and interleukin (IL)-2 in response to hematologic (Jeko-1, RPMI-8226) and solid (OVCAR-3, Capan-2, NCI-H226) tumor cell lines in vitro. In vivo, CAR-1 and CAR-2 both cleared hematologic Jeko-1 lymphoma xenografts, however only CAR-1 fully rejected ovarian solid OVCAR-3 tumors, concordantly with greater expansion of CD8+ and CD4+CAR T cells, and enrichment for central and effector memory phenotype. When equipped with TGFβ-protective armor TGFβRIIDN, CAR-1 T cells resisted TGFβ-mediated pSmad2/3 phosphorylation, as compared with CAR-1 alone. When co-cultured with ROR-1+ AsPC-1 pancreatic cancer line in the presence of TGFβ1, armored CAR-1 demonstrated improved recovery of killing function, IFN-γ, TNF-α and IL-2 secretion. In mouse AsPC-1 pancreatic tumor xenografts overexpressing TGFβ1, armored CAR-1, in contrast to CAR-1 alone, achieved complete tumor remissions, and yielded accelerated expansion of CAR+ T cells, diminished circulating active TGFβ1, and no apparent toxicity or weight loss. Unexpectedly, in AsPC-1 xenografts without TGFβ overexpression, TGFβ1 production was specifically induced by ROR-1-CAR T cells interaction with ROR-1 positive tumor cells, and the TGFβRIIDN armor conferred accelerated tumor clearance.
    CONCLUSIONS: The novel fully human TGFßRIIDN-armored ROR1-CAR-1 T cells are highly potent against ROR1-positive tumors, and withstand the inhibitory effects of TGFß in solid TME. Moreover, TGFβ1 induction represents a novel, CAR-induced checkpoint in the solid TME, which can be circumvented by co-expressing the TGβRIIDN armor 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
    背景:通过ROR1及其伴侣通过WNT5A进行的非规范WNT家族(WNT5A通路)信号传导,ROR2或Frizzled2(FZD2)与驱动肿瘤发生和治疗抵抗的过程有关。我们利用大量的尿路上皮癌(UC)肿瘤数据集来表征通过WNT5A的非规范WNT信号,ROR1、ROR2或FZD2表达。
    方法:NextGen对提交给CarisLifeSciences的4125个UC肿瘤进行DNA(592个基因或WES)/RNA(WTS)测序。WNT5A的高表达和低表达,ROR1,ROR2和FZD2被定义为≥上四分位数和<下四分位数/百万(TPM),分别。分析基因表达谱的转录特征,预测对免疫疗法的反应。在适当的情况下应用了Mann-WhitneyU和X2/Fisher精确检验,P值调整为多重比较(p<0.05)。从保险索赔数据获得真实世界总生存期(OS)。
    结果:WNT5A途径基因表达在原发与转移部位之间差异显著:WNT5A(25.2与16.8TPM),FZD2(3.2vs.4.05),ROR1(1.7vs.2.1),和ROR2(2.4与2.6)全部p<0.05。高表达和低表达亚组的比较显示TP53、FGFR3和RB1致病性突变的患病率存在差异。以及随着靶基因表达的增加,T细胞发炎的评分也增加。ROR2(HR1.31,95%CI1.15-1.50,p<0.001)和FZD2(HR1.16,95%CI1.02-1.32,p=0.024)的高基因表达与OS恶化相关。
    结论:在UC患者中观察到四种WNT5A通路成分的不同基因组和免疫景观。需要进行外部验证研究。
    BACKGROUND: Non-canonical WNT family (WNT5A pathway) signaling via WNT5A through ROR1 and its partner, ROR2, or Frizzled2 (FZD2) is linked to processes driving tumorigenesis and therapy resistance. We utilized a large dataset of urothelial carcinoma (UC) tumors to characterize non-canonical WNT signaling through WNT5A, ROR1, ROR2, or FZD2 expression.
    METHODS: NextGen Sequencing of DNA (592 genes or WES)/RNA (WTS) was performed for 4125 UC tumors submitted to Caris Life Sciences. High and low expression of WNT5A, ROR1, ROR2, and FZD2 was defined as ≥ top and RESULTS: WNT5A pathway gene expression varied significantly between primary versus metastatic sites: WNT5A (25.2 vs. 16.8 TPM), FZD2 (3.2 vs. 4.05), ROR1 (1.7 vs. 2.1), and ROR2 (2.4 vs. 2.6) p < 0.05 for all. Comparison of high- and low-expression subgroups revealed variation in the prevalence of TP53, FGFR3, and RB1 pathogenic mutations, as well as increasing T cell-inflamed scores as expression of the target gene increased. High gene expression for ROR2 (HR 1.31, 95% CI 1.15-1.50, p < 0.001) and FZD2 (HR 1.16, 95% CI 1.02-1.32, p = 0.024) was associated with worse OS.
    CONCLUSIONS: Distinct genomic and immune landscapes for the four investigated WNT5A pathway components were observed in patients with UC. External validation studies are needed.
    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
    目的:多模式治疗策略的可用性,包括靶向治疗和免疫疗法,改善了非小细胞肺癌(NSCLC)的生存率。然而,由于固有的抵抗力,一些患者仍然进展或反应不佳,获得性抵抗力,或者缺乏药物驱动突变。鞘氨醇-1-磷酸(S1P)和受体酪氨酸激酶样孤儿受体(ROR1/2)信号通路在肺癌发生过程中被激活。
    方法:在本研究中,我们已经评估了S1P和ROR1/2信号通路在肺癌细胞中的串扰。
    结果:S1P治疗肺癌细胞降低ROR1和ROR2转录水平。当用PF-543治疗时,一种药理学SphK1抑制剂或通过shRNA基因敲低SPHK1,引发ROR1和ROR2。此外,同时抑制SphK1和ROR1减少了肺癌细胞的迁移。
    结论:这些发现证明了两种途径的相互调节,这表明这两种途径都有反比关系,即在没有一条途径的情况下,另一条途径可能负责另一条途径。因此,同时靶向两种途径可作为肺癌治疗的潜在治疗靶点.
    OBJECTIVE: Availability of multimodal treatment strategies, including targeted therapies and immunotherapies, have improved the survival of non-small cell lung carcinoma (NSCLC). However, some patients still progress or respond poorly due to inherent resistance, acquired resistance, or lack of druggable driver mutations. Sphingosine-1-phosphate (S1P) and receptor tyrosine kinase-like orphan receptor (ROR1/2) signaling pathways are activated during lung carcinogenesis.
    METHODS: In this study, we have evaluated the crosstalk of S1P and ROR1/2 signaling pathways in lung cancer cells.
    RESULTS: S1P treatment of lung cancer cells decreases ROR1 and ROR2 transcript levels. While treatment with PF-543, a pharmacological SphK1 inhibitor or genetic knockdown of SPHK1 by shRNA, raises ROR1 and ROR2. Furthermore, simultaneous inhibition of SphK1 along with ROR1 reduced the migration of lung cancer cells.
    CONCLUSIONS: These findings demonstrate the reciprocal regulation of both pathways, suggesting that both pathways have an inverse relation i.e, in the absence of one pathway, another pathway may take charge of the other pathway. Therefore, simultaneously targeting both pathways could serve as a potential therapeutic target for lung cancer treatment.
    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细胞接合双特异性抗体已显示出血液恶性肿瘤概念的临床证据。尽管如此,细胞因子释放综合征,神经毒性,和上靶-外肿瘤毒性,尤其是在实体瘤环境中,是主要障碍。已经描述了第二代TCE,其通过降低对CD3和/或TAA的表观结合亲和力来使细胞毒性与细胞因子释放解耦,但是这种工程化的结果通常仅导致细胞因子释放的最大诱导减少,并且通常以最大细胞毒性为代价。使用ROR1作为我们的模型TAA和高度模块化的骆驼纳米体,我们描述了下一代解偶联TCE的工程改造,该TCE结合了一个\"细胞因子窗口\",定义为达到最大杀伤作用的剂量范围,但细胞因子释放可以从非常低的安全性调节到接近第一代TCE诱导的剂量范围.后一种属性支持包括旁观者杀伤在内的促炎抗肿瘤活性,并且可以被临床医生潜在地用于将患者剂量安全地滴定至认为比使用标准第二代方法可能更大的介导最大功效的剂量。我们使用了优化TCE介导的突触距离和TAA结合臂的表观亲和力调节的组合方法,以产生支持宽细胞因子窗口的相对较长但持久的突触。有效的杀伤和减少免疫衰竭的倾向。重要的是,这种下一代TCE在体内诱导了显着的肿瘤生长抑制,但与诱导致死性CRS的第一代非解偶联基准TCE不同,未观察到不良事件的迹象.
    T cell engaging bispecific antibodies have shown clinical proof of concept for hematologic malignancies. Still, cytokine release syndrome, neurotoxicity, and on-target-off-tumor toxicity, especially in the solid tumor setting, represent major obstacles. Second generation TCEs have been described that decouple cytotoxicity from cytokine release by reducing the apparent binding affinity for CD3 and/or the TAA but the results of such engineering have generally led only to reduced maximum induction of cytokine release and often at the expense of maximum cytotoxicity. Using ROR1 as our model TAA and highly modular camelid nanobodies, we describe the engineering of a next generation decoupled TCE that incorporates a \"cytokine window\" defined as a dose range in which maximal killing is reached but cytokine release may be modulated from very low for safety to nearly that induced by first generation TCEs. This latter attribute supports pro-inflammatory anti-tumor activity including bystander killing and can potentially be used by clinicians to safely titrate patient dose to that which mediates maximum efficacy that is postulated as greater than that possible using standard second generation approaches. We used a combined method of optimizing TCE mediated synaptic distance and apparent affinity tuning of the TAA binding arms to generate a relatively long but persistent synapse that supports a wide cytokine window, potent killing and a reduced propensity towards immune exhaustion. Importantly, this next generation TCE induced significant tumor growth inhibition in vivo but unlike a first-generation non-decoupled benchmark TCE that induced lethal CRS, no signs of adverse events were observed.
    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
    间皮单层的粘附和清除是卵巢癌转移种植的关键早期事件。ROR2是与Wnt5a配体相互作用以激活非经典Wnt信号传导的受体酪氨酸激酶,并且先前已显示在卵巢癌组织中上调。然而,之前没有研究评估ROR2在卵巢癌中的作用机制.通过细胞高通量遗传筛选,我们独立确定ROR2是卵巢肿瘤细胞粘附和侵袭的驱动因素.ROR2在卵巢肿瘤细胞中的表达用于驱动定向细胞优先向高Wnt5a配体区域迁移,如间皮衬里网膜。此外,ROR2促进卵巢肿瘤细胞粘附和间皮单层的清除。肿瘤细胞中ROR2的耗尽,减少同系卵巢癌模型中的转移性肿瘤负担。这些发现支持ROR2在卵巢肿瘤细胞中作为促成早期转移的关键因素的作用。ROR2/Wnt5a信号轴的治疗靶向可以提供改善晚期卵巢癌患者治疗的手段。含义:这项研究表明,卵巢癌细胞中的ROR2对于定向迁移到转移性生态位很重要,并为卵巢癌的治疗靶向提供了潜在的目标信号轴。
    Adhesion to and clearance of the mesothelial monolayer are key early events in metastatic seeding of ovarian cancer. ROR2 is a receptor tyrosine kinase that interacts with Wnt5a ligand to activate noncanonical Wnt signaling and has been previously shown to be upregulated in ovarian cancer tissue. However, no prior study has evaluated the mechanistic role of ROR2 in ovarian cancer. Through a cellular high-throughput genetic screen, we independently identified ROR2 as a driver of ovarian tumor cell adhesion and invasion. ROR2 expression in ovarian tumor cells serves to drive directed cell migration preferentially toward areas of high Wnt5a ligand, such as the mesothelial lined omentum. In addition, ROR2 promotes ovarian tumor cell adhesion and clearance of a mesothelial monolayer. Depletion of ROR2, in tumor cells, reduces metastatic tumor burden in a syngeneic model of ovarian cancer. These findings support the role of ROR2 in ovarian tumor cells as a critical factor contributing to the early steps of metastasis. Therapeutic targeting of the ROR2/Wnt5a signaling axis could provide a means of improving treatment for patients with advanced ovarian cancer.
    UNASSIGNED: This study demonstrates that ROR2 in ovarian cancer cells is important for directed migration to the metastatic niche and provides a potential signaling axis of interest for therapeutic targeting in ovarian cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号