MSS, microsatellite stable

  • 文章类型: Journal Article
    非洲(AFR)血统的个体的结直肠癌(CRC)发病率高于欧洲(EUR)血统的个体,并且表现出显著的健康差异。先前的研究已经注意到AFR和EURCRC患者之间的肿瘤微环境差异。然而,支持这些免疫差异的分子调控过程在很大程度上仍然未知。
    使用癌症基因组图谱对55名AFR和456名EUR患者进行了多组学分析。我们通过使用基因表达和甲基化数据评估肿瘤微环境,转录因子,和掌握转录调节子分析,以确定介导观察到的表型差异的细胞信号传导途径。
    我们证明AFR患者中下调的基因显示出典型途径的富集,包括趋化因子信号。此外,对肿瘤微环境的评估显示,与EUR患者相比,AFR中的细胞毒性淋巴细胞和中性粒细胞数量显着减少,提示AFR患者的免疫反应减弱。我们进一步证明,称为“主转录调节因子”(MTR)的分子在通过疾病相关转录因子(TF)介导的复杂信号转导网络调节影响关键免疫过程的基因表达中起关键作用。此外,一组核心的这些MTR和TF显示与AFR和EURCRC患者的细胞毒性淋巴细胞和中性粒细胞水平呈正相关,因此表明它们在驱动两个祖先群体之间的免疫浸润差异中的作用。
    我们的研究提供了对MTR和TF的复杂调控景观的见解,这些景观协调了AFR和EUR血统的CRC患者之间肿瘤微环境的差异。
    UNASSIGNED: Individuals of African (AFR) ancestry have a higher incidence of colorectal cancer (CRC) than those of European (EUR) ancestry and exhibit significant health disparities. Previous studies have noted differences in the tumor microenvironment between AFR and EUR patients with CRC. However, the molecular regulatory processes that underpin these immune differences remain largely unknown.
    UNASSIGNED: Multiomics analysis was carried out for 55 AFR and 456 EUR patients with microsatellite-stable CRC using The Cancer Genome Atlas. We evaluated the tumor microenvironment by using gene expression and methylation data, transcription factor, and master transcriptional regulator analysis to identify the cell signaling pathways mediating the observed phenotypic differences.
    UNASSIGNED: We demonstrate that downregulated genes in AFR patients with CRC showed enrichment for canonical pathways, including chemokine signaling. Moreover, evaluation of the tumor microenvironment showed that cytotoxic lymphocytes and neutrophil cell populations are significantly decreased in AFR compared with EUR patients, suggesting AFR patients have an attenuated immune response. We further demonstrate that molecules called \"master transcriptional regulators\" (MTRs) play a critical role in regulating the expression of genes impacting key immune processes through an intricate signal transduction network mediated by disease-associated transcription factors (TFs). Furthermore, a core set of these MTRs and TFs showed a positive correlation with levels of cytotoxic lymphocytes and neutrophils across both AFR and EUR patients with CRC, thus suggesting their role in driving the immune infiltrate differences between the two ancestral groups.
    UNASSIGNED: Our study provides an insight into the intricate regulatory landscape of MTRs and TFs that orchestrate the differences in the tumor microenvironment between patients with CRC of AFR and EUR ancestry.
    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
    结直肠癌(CRC),世界范围内的恶性肿瘤由微卫星不稳定性(MSI)和稳定(MSS)表型组成。尽管SHP2是癌症治疗的一个有希望的靶点,它与先天免疫抑制的关系仍然难以捉摸。为了解决这个问题,进行单细胞RNA测序以探索SHP2在小鼠MC38异种移植物的所有细胞类型的肿瘤微环境(TME)中的作用。发现瘤内细胞在功能上是异质的,并且对SHP2变构抑制剂SHP099有显着反应。SHP099明显阻止了肿瘤细胞的恶性演变。机械上,STING-TBK1-IRF3介导的I型干扰素信号在浸润的骨髓细胞中被SHP099高度激活。值得注意的是,与MSI高表型相比,具有MSS表型的CRC患者在CD68巨噬细胞中表现出更大的巨噬细胞浸润和更有效的SHP2磷酸化,提示巨噬细胞SHP2在TME中的潜在作用。总的来说,我们的数据揭示了SHP2介导的先天免疫抑制机制,提示SHP2是结肠癌免疫治疗的一个有前景的靶点.
    Colorectal cancer (CRC), a malignant tumor worldwide consists of microsatellite instability (MSI) and stable (MSS) phenotypes. Although SHP2 is a hopeful target for cancer therapy, its relationship with innate immunosuppression remains elusive. To address that, single-cell RNA sequencing was performed to explore the role of SHP2 in all cell types of tumor microenvironment (TME) from murine MC38 xenografts. Intratumoral cells were found to be functionally heterogeneous and responded significantly to SHP099, a SHP2 allosteric inhibitor. The malignant evolution of tumor cells was remarkably arrested by SHP099. Mechanistically, STING-TBK1-IRF3-mediated type I interferon signaling was highly activated by SHP099 in infiltrated myeloid cells. Notably, CRC patients with MSS phenotype exhibited greater macrophage infiltration and more potent SHP2 phosphorylation in CD68+ macrophages than MSI-high phenotypes, suggesting the potential role of macrophagic SHP2 in TME. Collectively, our data reveals a mechanism of innate immunosuppression mediated by SHP2, suggesting that SHP2 is a promising target for colon cancer immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    耐药性和缺乏分子治疗靶标是骨肉瘤(OS)管理的主要挑战。鉴定与OS复发和化疗抗性相关的新的遗传改变将具有科学和临床意义。
    为了确定与OS复发和化疗耐药相关的潜在遗传改变,我们收集了一名20岁男性骨肉瘤患者的原发灶(p-OS)和辅助化疗5个月后形成的转移性肿瘤(m-OS)的活检.两种OS标本均进行癌症靶向下一代测序(NGS),并在三维条件下培养其细胞悬液以建立球体治疗模型。针对检测到的突变基因GPC3的面向转录本的Sanger测序,在p-OS和m-OS组织和球状体的RNA样品上进行。阐明了抗GPC3抗体及其与顺铂的组合对m-OS球体的影响。
    NGS在p-OS中显示了4个突变(GPC3,SOX10,MDM4和MAPK8)和6个扩增(MDM2,CDK4,CCND3,RUNX2,GLI1和FRS2),和m-OS中的3个突变(GPC3,SOX10和EGF)和10个扩增(CDK4,CCND3,MDM2,RUNX2,GLI1,FRS2,CARD11,RAC1,SLC16A7和PMS2)。在这些改动中,GPC3的突变丰度在p-OS中最高(56.49%),在m-OS中增加1.54倍。面向GPC3转录本的Sanger测序证实了外显子4中1046处的突变,免疫组织化学染色显示m-OS组织及其球状体中GPC3的产生增加。EdU细胞增殖和钙黄绿素/PI细胞活力测定揭示了抗OS一线药物(多柔比星,顺铂,甲氨蝶呤,异环磷酰胺和卡铂),10μM卡铂对p-OS的抑制作用最好,但对m-OS球体没有抑制作用。2μg/mL抗GPC3抗体有效地使m-OS球体自行死亡(76.43%)或与顺铂联合使用(92.93%)。
    这项研究表明,在转移性骨肉瘤及其具有多药耐药性的球体中,突变型GPC3的丰度增加和表达上调。由于GPC3靶向治疗已用于治疗肝细胞癌,并且对OSPDS也有效,GPC3可能是骨肉瘤的一个新的预后参数和治疗靶点。
    UNASSIGNED: Drug resistance and the lack of molecular therapeutic target are the main challenges in the management of osteosarcomas (OSs). Identification of novel genetic alteration(s) related with OS recurrence and chemotherapeutic resistance would be of scientific and clinical significance.
    UNASSIGNED: To identify potential genetic alterations related with OS recurrence and chemotherapeutic resistance, the biopsies of a 20-year-old male osteosarcoma patient were collected at primary site (p-OS) and from its metastatic tumor (m-OS) formed after 5 months of adjuvant chemotherapy. Both OS specimens were subjected to cancer-targeted next generation sequencing (NGS) and their cell suspensions were cultured under three-dimensional condition to establish spheroid therapeutic model. Transcript-oriented Sanger sequencing for GPC3, the detected mutated gene, was performed on RNA samples of p-OS and m-OS tissues and spheroids. The effects of anti-GPC3 antibody and its combination with cisplatin on m-OS spheroids were elucidated.
    UNASSIGNED: NGS revealed 4 mutations (GPC3, SOX10, MDM4 and MAPK8) and 6 amplifications (MDM2, CDK4, CCND3, RUNX2, GLI1 and FRS2) in p-OS, and 3 mutations (GPC3, SOX10 and EGF) and 10 amplifications (CDK4, CCND3, MDM2, RUNX2, GLI1, FRS2, CARD11, RAC1, SLC16A7 and PMS2) in m-OS. Among those alterations, the mutation abundance of GPC3 was the highest (56.49%) in p-OS and showed 1.54 times increase in m-OS. GPC3 transcript-oriented Sanger sequencing confirmed the mutation at 1046 in Exon 4, and immunohistochemical staining showed increased GPC3 production in m-OS tissues and its spheroids. EdU cell proliferation and Calcein/PI cell viability assays revealed that of the anti-OS first line drugs (doxorubicin, cisplatin, methotrexate, ifosfamide and carboplatin), 10 μM carboplatin exerted the best inhibitory effects on the p-OS but not the m-OS spheroids. 2 μg/mL anti-GPC3 antibody effectively committed m-OS spheroids to death by itself (76.43%) or in combination with cisplatin (92.93%).
    UNASSIGNED: This study demonstrates increased abundance and up-regulated expression of mutant GPC3 in metastatic osteosarcoma and its spheroids with multidrug resistance. As GPC3-targeting therapy has been used to treat hepatocellular carcinomas and it is also effective to OS PDSs, GPC3 would be a novel prognostic parameter and therapeutic target of osteosarcomas.
    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
    Advancement in the field of cancer genomics is revolutionizing the molecular characterization of a wide variety of different cancers. Recent application of large-scale, next-generation sequencing technology to gastric cancer, which remains a major source of morbidity and mortality throughout the world, has helped better define the complex genomic landscape of this cancer. These studies also have led to the development of novel genomically based molecular classification systems for gastric cancer, reinforced the importance of classic driver mutations in gastric cancer pathogenesis, and led to the discovery of new driver gene mutations that previously were not known to be associated with gastric cancer. This wealth of genomic data has significant potential to impact the future management of this disease, and the challenge remains to effectively translate this genomic data into better treatment paradigms for gastric cancer.
    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
    Although tumor infiltrating lymphocyte (TIL) density is prognostic and predictive in colorectal cancer (CRC), the impact of tumor genetics upon colorectal immunobiology is unclear. Identification of genetic factors that influence the tumor immunophenotype is essential to improve the effectiveness of stratified immunotherapy approaches. We carried out a bioinformatics analysis of CRC data in The Cancer Genome Atlas (TCGA) involving two-dimensional hierarchical clustering to define an immune signature that we used to characterize the immune response across key patient groups. An immune signature termed The Co-ordinate Immune Response Cluster (CIRC) comprising 28 genes was coordinately regulated across the patient population. Four patient groups were delineated on the basis of cluster expression. Group A, which was heavily enriched for patients with microsatellite instability (MSI-H) and POL mutations, exhibited high CIRC expression, including the presence of several inhibitory molecules: CTLA4, PDL1, PDL2, LAG3, and TIM3. In contrast, RAS mutation was enriched in patient groups with lower CIRC expression. This work links the genetics and immunobiology of colorectal tumorigenesis, with implications for the development of stratified immunotherapeutic approaches. Microsatellite instability and POL mutations are linked with high mutational burden and high immune infiltration, but the coordinate expression of inhibitory pathways observed suggests combination checkpoint blockade therapy may be required to improve efficacy. In contrast, RAS mutant tumors predict for a relatively poor immune infiltration and low inhibitory molecule expression. In this setting, checkpoint blockade may be less efficacious, highlighting a requirement for novel strategies in this patient group.
    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
    The CpG Island Methylator Phenotype (CIMP) is fundamental to an important subset of colorectal cancer; however, its cause is unknown. CIMP is associated with microsatellite instability but is also found in BRAF mutant microsatellite stable cancers that are associated with poor prognosis. The isocitrate dehydrogenase 1 (IDH1) gene causes CIMP in glioma due to an activating mutation that produces the 2-hydroxyglutarate oncometabolite. We therefore examined IDH1 alteration as a potential cause of CIMP in colorectal cancer. The IDH1 mutational hotspot was screened in 86 CIMP-positive and 80 CIMP-negative cancers. The entire coding sequence was examined in 81 CIMP-positive colorectal cancers. Forty-seven cancers varying by CIMP-status and IDH1 mutation status were examined using Illumina 450K DNA methylation microarrays. The R132C IDH1 mutation was detected in 4/166 cancers. All IDH1 mutations were in CIMP cancers that were BRAF mutant and microsatellite stable (4/45, 8.9%). Unsupervised hierarchical cluster analysis identified an IDH1 mutation-like methylation signature in approximately half of the CIMP-positive cancers. IDH1 mutation appears to cause CIMP in a small proportion of BRAF mutant, microsatellite stable colorectal cancers. This study provides a precedent that a single gene mutation may cause CIMP in colorectal cancer, and that this will be associated with a specific epigenetic signature and clinicopathological features.
    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