Carbohydrate sulfotransferase 15 (CHST15)

  • 文章类型: Journal Article
    未经证实:基质靶向治疗对肿瘤免疫抑制的影响在很大程度上尚未被研究。RNA寡核苷酸,STNM01已显示抑制负责肿瘤蛋白聚糖合成和基质重塑的碳水化合物磺基转移酶15(CHST15)。这项I/IIa期研究旨在评估STNM01在不可切除的胰腺导管腺癌(PDAC)患者中的安全性和有效性。
    未经批准:这是一个开放标签,STNM01作为吉西他滨联合nab-紫杉醇难治性PDAC二线治疗的剂量递增研究.一个周期包括三次2周内窥镜超声引导的局部注射STNM01,剂量为250、1,000、2,500或10,000nM,与S-1组合(每天两次80-120mg,每3周14天)。主要结果是剂量-剂量毒性(DLT)的发生率。次要结局包括总生存期(OS),肿瘤反应,肿瘤微环境的变化对免疫组织病理学的影响,和安全性(jRCT2031190055)。
    未经批准:共纳入22例患者,最多重复3个循环;未观察到DLT。中位OS为7.8个月。疾病控制率为77.3%;1例患者显示胰腺和肿瘤引流淋巴结可见病变完全消失。较高的肿瘤CHST15表达与基线时较差的CD3+和CD8+T细胞浸润相关。STNM01导致CHST15的显着减少,并在第1周期结束时与S-1组合的肿瘤浸润性CD3和CD8T细胞增加。CD3+T细胞中更高的倍数增加与更长的OS相关。有8个3级不良事件。
    UNASSIGNED:局部注射STNM01作为联合二线治疗的不可切除PDAC患者的耐受性良好。它通过增强肿瘤微环境中的T细胞浸润来延长存活。
    UNASSIGNED:本研究得到了日本医学研究与发展机构(AMED)的支持。
    UNASSIGNED: The impact of stroma-targeting therapy on tumor immune suppression is largely unexplored. An RNA oligonucleotide, STNM01, has been shown to repress carbohydrate sulfotransferase 15 (CHST15) responsible for tumor proteoglycan synthesis and matrix remodeling. This phase I/IIa study aimed to evaluate the safety and efficacy of STNM01 in patients with unresectable pancreatic ductal adenocarcinoma (PDAC).
    UNASSIGNED: This was an open-label, dose-escalation study of STNM01 as second-line therapy in gemcitabine plus nab-paclitaxel-refractory PDAC. A cycle comprised three 2-weekly endoscopic ultrasound-guided locoregional injections of STNM01 at doses of 250, 1,000, 2,500, or 10,000 nM in combination with S-1 (80-120 mg twice a day for 14 days every 3 weeks). The primary outcome was the incidence of dose-liming toxicity (DLT). The secondary outcomes included overall survival (OS), tumor response, changes in tumor microenvironment on immunohistopathology, and safety (jRCT2031190055).
    UNASSIGNED: A total of 22 patients were enrolled, and 3 cycles were repeated at maximum; no DLT was observed. The median OS was 7.8 months. The disease control rate was 77.3%; 1 patient showed complete disappearance of visible lesions in the pancreas and tumor-draining lymph nodes. Higher tumoral CHST15 expression was associated with poor CD3+ and CD8+ T cell infiltration at baseline. STNM01 led to a significant reduction in CHST15, and increased tumor-infiltrating CD3+ and CD8+ T cells in combination with S-1 at the end of cycle 1. Higher fold increase in CD3+ T cells correlated with longer OS. There were 8 grade 3 adverse events.
    UNASSIGNED: Locoregional injection of STNM01 was well tolerated in patients with unresectable PDAC as combined second-line therapy. It prolonged survival by enhancing T cell infiltration in tumor microenvironment.
    UNASSIGNED: The present study was supported by the Japan Agency for Medical Research and Development (AMED).
    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
    未经证实:腹膜后脂肪肉瘤(RPLS)是一种罕见的,具有独特临床特征的生物异质性肿瘤,例如频繁的局部复发,反复复发,和罕见的远处转移。RPLS没有有效的靶向治疗。这里,我们的目的是确定RPLS中碳水化合物磺基转移酶15(CHST15)的病理功能和治疗潜力。
    UNASSIGNED:肿瘤来源的间充质祖细胞(MPCs)和正常脂肪来源的间充质干细胞(MSCs)来自RPLS患者。分离MPCs和MSCs,并根据表面标记进行表征,扩散,并使用流式细胞术和分子染色进行分化。进行转录组分析以破译3个配对MSC和MPC中的分化相关基因的表达谱。使用定量实时聚合酶链反应(qRT-PCR)进行基因的进一步确认。将过表达CHST15的质粒转染到脂肪MSC中以通过实时PCR检查mRNA水平的纤维化相关基因表达。
    UNASSIGNED:肿瘤基质来源的MPCs表达CD105、CD73和CD90,并在体外显示成骨和成脂分化潜能。肿瘤来源的MPCs的增殖明显低于正常脂肪来源的MSCs(P<0.001)。转录组分析显示IL-7R上调,ALPL,肿瘤源性MPCs中的PKNOX2和CHST15。CHST15在肿瘤来源的MPCs中高表达(P<0.001)。CHST15介导的纤维化相关FGF2基因在MSCs(P<0.05)和MPCs(P<0.001)中的表达。
    未经证实:CHST15在肿瘤衍生的MPC中上调,并在RPLS中调节纤维化。这为通过靶向CHST15诱导的RPLS中的MPC激活开发新的治疗策略提供了线索。
    UNASSIGNED: Retroperitoneal liposarcoma (RPLS) is a rare, biologically heterogeneous tumor with distinct clinical characteristics, such as frequent local recurrence, repeated relapse, and rare distant metastasis. No effective targeted therapy is available for RPLS. Here, we aim to determine the pathological functions and therapeutic potential of carbohydrate sulfotransferase 15 (CHST15) in RPLS.
    UNASSIGNED: Tumor-derived mesenchymal progenitor cells (MPCs) and normal adipose derived mesenchymal stem cells (MSCs) were obtained from patients with RPLS. MPCs and MSCs were isolated and characterized based on surface markers, proliferation, and differentiation using flow cytometry and molecular staining. Transcriptome analysis was performed to decipher expression profile of differentiation-related genes in 3 paired MSCs and MPCs. Further confirmation of genes were performed using quantitative real-time polymerase chain reaction (qRT-PCR). Plasmids overexpressing CHST15 were transfected into adipose MSCs to examine fibrosis-related gene expression at mRNA level by real-time PCR.
    UNASSIGNED: The tumor stromal-derived MPCs expressed CD105, CD73, and CD90, and exhibited osteogenic and adipogenic differentiation potential in vitro. The proliferation of tumor-derived MPCs was significantly lower than that of normal adipose-derived MSCs (P<0.001). Transcriptome analysis revealed upregulation of IL-7R, ALPL, PKNOX2, and CHST15 in tumor-derived MPCs. CHST15 was highly expressed in tumor-derived MPCs (P<0.001). CHST15 mediated fibrosis-related FGF2 gene expression in MSCs (P<0.05) and MPCs (P<0.001).
    UNASSIGNED: CHST15 is upregulated in tumor-derived MPCs and regulates fibrosis in RPLS. This provides clues for development of novel therapeutic strategies by targeting CHST15-induced MPC activation in RPLS.
    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
    Fibrosis continues to be paid a great attention in not only basic research but also clinical practice, especially for the development of novel therapeutics in various fibrotic diseases. However, there remain several obstacles to translation in developing anti-fibrosis therapy. The present review documents our translational practice from target discovery to first-in-patient studies in the development of anti-fibrosis therapy for inflammatory bowel disease (IBD). First topic is a target selection. We have focused on the target that has an ability to regulate multifactorial cascades of fibrosis. Carbohydrate sulfotransferase 15 (CHST15) synthesizes matrix proteoglycan that regulates various pathogenic mediators and contributes to tissue remodeling during injury. Small interfering RNA (siRNA) targeting CHST15 inhibited activation of fibroblasts in vitro and reduced fibrosis in vivo. Second topic is a clinically feasible application. We established a safe and novel pancolonic delivery of siRNA, which is achieved by direct injection to extracellular matrix (ECM) through endoscope. Third topic is an endpoint for both nonclinical and clinical studies. We have focused on tissue-specific findings for co-existence of fibrosis in ulcerative lesions in IBD and investigated whether the balance of mucosal healing (MH) and fibrosis, which is evaluated by endoscopy and histology respectively, can be used for study endpoints. Phase 1 clinical trial of STNM01, a synthesized CHST15 siRNA, by a single dose endoscopic submucosal injection for non-healer patients with Crohn\'s disease showed high rates of MH. Analyses of biopsy specimens revealed that STNM01 reduced CHST15 expression at local lesions, repressed pre-existing fibrosis and repaired the damaged crypts. Thus, blockade of multifactorial modulator CHST15 in ECM showed a potential to treat tissue remodeling and skew fibrosis toward mucosal repair. Our practice suggests that target- and tissue-specific findings-based strategy would be a key to translation in developing anti-fibrosis therapy.
    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