解密胃癌腹膜转移的转移途径特异性信号及其微环境相互作用

IF 20.1 1区 医学 Q1 ONCOLOGY Cancer Communications Pub Date : 2024-03-18 DOI:10.1002/cac2.12533
Ki Tae Kim, Jae Eun Lee, Jae-Ho Cheong, In Cho, Yoon Young Choi
{"title":"解密胃癌腹膜转移的转移途径特异性信号及其微环境相互作用","authors":"Ki Tae Kim,&nbsp;Jae Eun Lee,&nbsp;Jae-Ho Cheong,&nbsp;In Cho,&nbsp;Yoon Young Choi","doi":"10.1002/cac2.12533","DOIUrl":null,"url":null,"abstract":"<p>Gastric cancer (GC) is a pervasive global malignancy with high mortality rates due to distant metastasis [<span>1</span>]. GC metastasis can occur via hematogenous route, peritoneal route, and specifically through ovarian spread in females [<span>2</span>]. Among these, peritoneal metastasis is the most prevalent and challenging condition to treat [<span>3</span>]. The Cancer Genome Atlas (TCGA) has uncovered four molecular subtypes of GC: microsatellite instability, Epstein-Barr virus-related, chromosomal instability, and genomically stable [<span>4</span>]. These subtypes exhibit unique features and metastatic behaviors [<span>5</span>], providing valuable insights into the molecular characteristics of GC. Nevertheless, there remains a gap in understanding gene expression feature in metastatic GC compared to corresponding primary tumors, particularly regarding its biological and clinical relevance in different metastatic patterns.</p><p>A whole-transcriptome sequencing analysis was conducted to investigate the characteristics of metastatic GC. We examined 66 paired primary and metastatic tumors, categorized according to their sites (38 primary, 9 hematogenous, 6 peritoneal, and 13 ovarian metastases), collected from 14 patients with GC metastasis (Supplementary Tables S1-S2 and Supplementary Figure S1). To corroborate the biological and clinical relevance of the identified metastatic GC-specific signals, we utilized publicly available databases containing expression data on primary GC, including TCGA (<i>n</i> = 415) [<span>4</span>], Asian Cancer Research Group (ACRG, <i>n</i> = 300) [<span>6</span>], and Yonsei Cancer Center (YCC, <i>n</i> = 433) cohorts [<span>7</span>]. For in-depth analysis at the single-cell resolution, we accessed a public dataset comprising 5 primary GC samples [<span>8</span>] and performed a single-cell RNA sequencing (scRNA-seq) analysis on ascites obtained from 4 patients with GC (Supplementary Tables S3-S4). The flow of analysis is summarized in Supplementary Figure S2. Details of the patient demographics, samples, and data analysis methods used are outlined in the Supplementary Methods.</p><p>Through a differential expression analysis of the 66 paired primary and metastatic tumors, we identified 949 genes uniquely associated with metastatic GC tumors (Supplementary Figure S3). Subsequent over-representation analysis of these metastatic GC-specific genes indicated that hematogenous metastatic tumors exhibited an up-regulation of receptor tyrosine kinase-related signals, while simultaneously showing a down-regulation of pathways associated with epithelial-mesenchymal transition (EMT) (Supplementary Figure S4). Conversely, peritoneal/ovarian metastatic tumors demonstrated an upregulation of the Hedgehog pathway, a key player in EMT signaling. These findings suggest that the mechanisms underlying GC metastasis vary by route [<span>5</span>], thus underscoring the need for route-specific clinical considerations.</p><p>To assess the clinical and biological significance of the identified genes, we examined their expression profiles in the TCGA cohort, which consisted of 415 primary GC tumors [<span>4</span>]. The non-negative factorization (NMF) algorithm stratified TCGA cohort into two subtypes based on the expression of GC-specific genes, denoted as NMF-G1 and NMF-G2. The NMF-G2 subtype, characterized by upregulated expression of GC-specific genes, was associated with the genomically stable subtype. It frequently exhibited somatic mutations in the cadherin 1 (<i>CDH1</i>) and was associated with a poor prognosis, regardless of the molecular subtypes (Supplementary Figure S5). Through weighted gene co-expression network analysis, we identified a module comprising 122 genes with distinctive expression patterns between NMF-G1 and NMF-G2. Notably, these genes exhibited the upregulation of EMT-related signals, leading to their designation as metastasis-specific EMT (msEMT) genes (Supplementary Figure S6). These msEMT genes are unique as they differ from established gene sets in GC biology and consensus cancer driver genes [<span>6, 9</span>]. Furthermore, the majority of msEMT genes exhibited downregulation in hematogenous metastasis. Upon re-evaluation, the expression level of these genes was higher in peritoneal/ovarian metastatic tumors than in corresponding primary tumors (Supplementary Tables S5-S6).</p><p>To validate the clinical relevance of msEMT genes in GC, we analyzed their expression levels in two independent primary GC cohorts from ACRG [<span>6</span>] and YCC [<span>7</span>], which had recurrence data. Assessment of the msEMT gene expression profile enabled the classification of each cohort into two distinct subtypes, with the high-expression group designated as the msEMT subtype. Importantly, the msEMT subtype was associated with a significantly worse prognosis in both cohorts, even after adjusting for clinical factors (Supplementary Figure S7A-D). Furthermore, the msEMT subtype in primary tumors was more closely linked to peritoneal/ovarian recurrence. Machine learning analyses showed that msEMT genes had a strong predictive capacity for peritoneal/ovarian metastasis, with high accuracy (&gt;0.80), particularly for negative predictive values and specificity, compared with models involving clinical variables (Supplementary Figure S7E and Supplementary Table S7). These results highlight the pivotal role of msEMT genes in the occurrence of peritoneal/ovarian metastasis and suggest that primary GC possesses metastatic potential even before the onset of peritoneal/ovarian metastasis; therefore, msEMT genes could serve as potential biomarkers for risk assessment.</p><p>To understand the origin of msEMT gene expression in GC, we conducted a scRNA-seq analysis of primary tumors (stratified by superficial and deep layer) [<span>8</span>] and ascites from 4 patients with gastric cancer metastasis. msEMT genes were not predominantly expressed in cancer cells, but rather in fibroblasts within the deep layers of primary tumors and in ascites (Supplementary Figure S8). This result indicated that msEMT signals are likely to arise from the tumor microenvironment, particularly from cancer-associated fibroblasts (CAFs), during the late stages of primary tumor development and peritoneal metastasis.</p><p>To further understand the expression patterns of msEMT genes in specific fibroblast populations, we explored their landscape and dynamic changes in fibroblasts. Within the deep layer of primary tumors, fibroblast lineage trajectory inference revealed that classical myofibroblastic CAFs (myo-CAFs, C1) and inflammatory CAFs (i-CAFs, C0) initiated the pseudo-time trajectory, whereas CAFs associated with cancer migration and EMT (C2) were at the end of the trajectory. msEMT genes exhibited differential expression across these fibroblast subtypes (Supplementary Figure S9A). In the ascites, vascular and inflammatory CAFs (C1 and C3) initiated the pseudo-time trajectory, myo-CAFs (C5) were positioned in the middle of the trajectory, and the mature CAFs associated with CAF proliferation and cancer cell migration (C6 and C7) concluded the trajectory (Supplementary Figure S9B). msEMT genes were mainly expressed across specific fibroblast subtypes, excepts myo-CAFs (C5). This implies their involvement in modulating the cellular landscape of fibroblasts during GC peritoneal metastasis.</p><p>When the gene expression profiles of fibroblasts within primary tumors were compared with those in ascites, we observed distinctly upregulated signals of extracellular matrix modulation in primary tumors, whereas ascites exhibited the upregulation of signals associated with cell-cell adhesion and immune response (Supplementary Figure S10). The interactions between fibroblasts and other cellular components are enhanced in ascites than in primary tumors, emphasizing the importance of comprehending cell-to-cell interactions in these contexts.</p><p>Cell-to-cell interaction analysis revealed that fibroblasts orchestrated and responded to signals associated with tumor growth and progression in primary tumors (Supplementary Figure S11A). Conversely, the fibroblasts within ascites predominantly receive signals associated with the modulation of angiogenesis and immune evasion, such as tumor necrosis factor-related weak inducer of apoptosis, complement, and macrophage migration inhibitory factor (Supplementary Figure S11B). The changes in fibroblast signaling reflect the alterations in the dynamic interplay between cancer cells and fibroblasts throughout the metastatic cascade, highlighting their pivotal role in the peritoneal dissemination of GC.</p><p>Our study has delineated the metastatic route-specific molecular profiles of GC, highlighting the prognostic impact of msEMT genes and the crucial role of CAFs in GC progression and peritoneal spread (Figure 1). These insights enhance our understanding of GC molecular variations by metastatic route and underscore the contribution of the tumor microenvironment to the dissemination of GC.</p><p>This study was approved by the Institutional Review Board of Severance Hospital (4-2019-0188) and Soonchunhyang University Bucheon Hospital (2022-09-008), and informed consent was received from all patients.</p>","PeriodicalId":9495,"journal":{"name":"Cancer Communications","volume":"44 4","pages":"514-517"},"PeriodicalIF":20.1000,"publicationDate":"2024-03-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12533","citationCount":"0","resultStr":"{\"title\":\"Deciphering metastatic route-specific signals and their microenvironment interactions in peritoneal metastasis of gastric cancer\",\"authors\":\"Ki Tae Kim,&nbsp;Jae Eun Lee,&nbsp;Jae-Ho Cheong,&nbsp;In Cho,&nbsp;Yoon Young Choi\",\"doi\":\"10.1002/cac2.12533\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p>Gastric cancer (GC) is a pervasive global malignancy with high mortality rates due to distant metastasis [<span>1</span>]. GC metastasis can occur via hematogenous route, peritoneal route, and specifically through ovarian spread in females [<span>2</span>]. Among these, peritoneal metastasis is the most prevalent and challenging condition to treat [<span>3</span>]. The Cancer Genome Atlas (TCGA) has uncovered four molecular subtypes of GC: microsatellite instability, Epstein-Barr virus-related, chromosomal instability, and genomically stable [<span>4</span>]. These subtypes exhibit unique features and metastatic behaviors [<span>5</span>], providing valuable insights into the molecular characteristics of GC. Nevertheless, there remains a gap in understanding gene expression feature in metastatic GC compared to corresponding primary tumors, particularly regarding its biological and clinical relevance in different metastatic patterns.</p><p>A whole-transcriptome sequencing analysis was conducted to investigate the characteristics of metastatic GC. We examined 66 paired primary and metastatic tumors, categorized according to their sites (38 primary, 9 hematogenous, 6 peritoneal, and 13 ovarian metastases), collected from 14 patients with GC metastasis (Supplementary Tables S1-S2 and Supplementary Figure S1). To corroborate the biological and clinical relevance of the identified metastatic GC-specific signals, we utilized publicly available databases containing expression data on primary GC, including TCGA (<i>n</i> = 415) [<span>4</span>], Asian Cancer Research Group (ACRG, <i>n</i> = 300) [<span>6</span>], and Yonsei Cancer Center (YCC, <i>n</i> = 433) cohorts [<span>7</span>]. For in-depth analysis at the single-cell resolution, we accessed a public dataset comprising 5 primary GC samples [<span>8</span>] and performed a single-cell RNA sequencing (scRNA-seq) analysis on ascites obtained from 4 patients with GC (Supplementary Tables S3-S4). The flow of analysis is summarized in Supplementary Figure S2. Details of the patient demographics, samples, and data analysis methods used are outlined in the Supplementary Methods.</p><p>Through a differential expression analysis of the 66 paired primary and metastatic tumors, we identified 949 genes uniquely associated with metastatic GC tumors (Supplementary Figure S3). Subsequent over-representation analysis of these metastatic GC-specific genes indicated that hematogenous metastatic tumors exhibited an up-regulation of receptor tyrosine kinase-related signals, while simultaneously showing a down-regulation of pathways associated with epithelial-mesenchymal transition (EMT) (Supplementary Figure S4). Conversely, peritoneal/ovarian metastatic tumors demonstrated an upregulation of the Hedgehog pathway, a key player in EMT signaling. These findings suggest that the mechanisms underlying GC metastasis vary by route [<span>5</span>], thus underscoring the need for route-specific clinical considerations.</p><p>To assess the clinical and biological significance of the identified genes, we examined their expression profiles in the TCGA cohort, which consisted of 415 primary GC tumors [<span>4</span>]. The non-negative factorization (NMF) algorithm stratified TCGA cohort into two subtypes based on the expression of GC-specific genes, denoted as NMF-G1 and NMF-G2. The NMF-G2 subtype, characterized by upregulated expression of GC-specific genes, was associated with the genomically stable subtype. It frequently exhibited somatic mutations in the cadherin 1 (<i>CDH1</i>) and was associated with a poor prognosis, regardless of the molecular subtypes (Supplementary Figure S5). Through weighted gene co-expression network analysis, we identified a module comprising 122 genes with distinctive expression patterns between NMF-G1 and NMF-G2. Notably, these genes exhibited the upregulation of EMT-related signals, leading to their designation as metastasis-specific EMT (msEMT) genes (Supplementary Figure S6). These msEMT genes are unique as they differ from established gene sets in GC biology and consensus cancer driver genes [<span>6, 9</span>]. Furthermore, the majority of msEMT genes exhibited downregulation in hematogenous metastasis. Upon re-evaluation, the expression level of these genes was higher in peritoneal/ovarian metastatic tumors than in corresponding primary tumors (Supplementary Tables S5-S6).</p><p>To validate the clinical relevance of msEMT genes in GC, we analyzed their expression levels in two independent primary GC cohorts from ACRG [<span>6</span>] and YCC [<span>7</span>], which had recurrence data. Assessment of the msEMT gene expression profile enabled the classification of each cohort into two distinct subtypes, with the high-expression group designated as the msEMT subtype. Importantly, the msEMT subtype was associated with a significantly worse prognosis in both cohorts, even after adjusting for clinical factors (Supplementary Figure S7A-D). Furthermore, the msEMT subtype in primary tumors was more closely linked to peritoneal/ovarian recurrence. Machine learning analyses showed that msEMT genes had a strong predictive capacity for peritoneal/ovarian metastasis, with high accuracy (&gt;0.80), particularly for negative predictive values and specificity, compared with models involving clinical variables (Supplementary Figure S7E and Supplementary Table S7). These results highlight the pivotal role of msEMT genes in the occurrence of peritoneal/ovarian metastasis and suggest that primary GC possesses metastatic potential even before the onset of peritoneal/ovarian metastasis; therefore, msEMT genes could serve as potential biomarkers for risk assessment.</p><p>To understand the origin of msEMT gene expression in GC, we conducted a scRNA-seq analysis of primary tumors (stratified by superficial and deep layer) [<span>8</span>] and ascites from 4 patients with gastric cancer metastasis. msEMT genes were not predominantly expressed in cancer cells, but rather in fibroblasts within the deep layers of primary tumors and in ascites (Supplementary Figure S8). This result indicated that msEMT signals are likely to arise from the tumor microenvironment, particularly from cancer-associated fibroblasts (CAFs), during the late stages of primary tumor development and peritoneal metastasis.</p><p>To further understand the expression patterns of msEMT genes in specific fibroblast populations, we explored their landscape and dynamic changes in fibroblasts. Within the deep layer of primary tumors, fibroblast lineage trajectory inference revealed that classical myofibroblastic CAFs (myo-CAFs, C1) and inflammatory CAFs (i-CAFs, C0) initiated the pseudo-time trajectory, whereas CAFs associated with cancer migration and EMT (C2) were at the end of the trajectory. msEMT genes exhibited differential expression across these fibroblast subtypes (Supplementary Figure S9A). In the ascites, vascular and inflammatory CAFs (C1 and C3) initiated the pseudo-time trajectory, myo-CAFs (C5) were positioned in the middle of the trajectory, and the mature CAFs associated with CAF proliferation and cancer cell migration (C6 and C7) concluded the trajectory (Supplementary Figure S9B). msEMT genes were mainly expressed across specific fibroblast subtypes, excepts myo-CAFs (C5). This implies their involvement in modulating the cellular landscape of fibroblasts during GC peritoneal metastasis.</p><p>When the gene expression profiles of fibroblasts within primary tumors were compared with those in ascites, we observed distinctly upregulated signals of extracellular matrix modulation in primary tumors, whereas ascites exhibited the upregulation of signals associated with cell-cell adhesion and immune response (Supplementary Figure S10). The interactions between fibroblasts and other cellular components are enhanced in ascites than in primary tumors, emphasizing the importance of comprehending cell-to-cell interactions in these contexts.</p><p>Cell-to-cell interaction analysis revealed that fibroblasts orchestrated and responded to signals associated with tumor growth and progression in primary tumors (Supplementary Figure S11A). Conversely, the fibroblasts within ascites predominantly receive signals associated with the modulation of angiogenesis and immune evasion, such as tumor necrosis factor-related weak inducer of apoptosis, complement, and macrophage migration inhibitory factor (Supplementary Figure S11B). The changes in fibroblast signaling reflect the alterations in the dynamic interplay between cancer cells and fibroblasts throughout the metastatic cascade, highlighting their pivotal role in the peritoneal dissemination of GC.</p><p>Our study has delineated the metastatic route-specific molecular profiles of GC, highlighting the prognostic impact of msEMT genes and the crucial role of CAFs in GC progression and peritoneal spread (Figure 1). These insights enhance our understanding of GC molecular variations by metastatic route and underscore the contribution of the tumor microenvironment to the dissemination of GC.</p><p>This study was approved by the Institutional Review Board of Severance Hospital (4-2019-0188) and Soonchunhyang University Bucheon Hospital (2022-09-008), and informed consent was received from all patients.</p>\",\"PeriodicalId\":9495,\"journal\":{\"name\":\"Cancer Communications\",\"volume\":\"44 4\",\"pages\":\"514-517\"},\"PeriodicalIF\":20.1000,\"publicationDate\":\"2024-03-18\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cac2.12533\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Cancer Communications\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/cac2.12533\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"ONCOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer Communications","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cac2.12533","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

胃癌(GC)是一种普遍存在的全球性恶性肿瘤,其远处转移导致的死亡率很高[1]。胃癌转移可通过血源性途径、腹膜途径,特别是通过女性卵巢转移[2]。其中,腹膜转移最为常见,治疗起来也最具挑战性[3]。癌症基因组图谱(TCGA)发现了 GC 的四种分子亚型:微卫星不稳定型、Epstein-Barr 病毒相关型、染色体不稳定型和基因组稳定型 [4]。这些亚型表现出独特的特征和转移行为[5],为了解 GC 的分子特征提供了宝贵的信息。然而,与相应的原发肿瘤相比,人们对转移性 GC 基因表达特征的了解仍然存在差距,尤其是在不同转移模式下的生物学和临床相关性方面。我们研究了从 14 例 GC 转移患者中收集的 66 例配对原发和转移肿瘤,并根据其部位进行了分类(38 例原发、9 例血行转移、6 例腹膜转移和 13 例卵巢转移)(补充表 S1-S2 和补充图 S1)。为了证实所发现的转移性 GC 特异性信号的生物学和临床相关性,我们利用了包含原发性 GC 表达数据的公开数据库,包括 TCGA(n = 415)[4]、亚洲癌症研究组(ACRG,n = 300)[6]和延世癌症中心(YCC,n = 433)队列[7]。为了在单细胞分辨率上进行深入分析,我们访问了由 5 个原发性 GC 样本组成的公开数据集[8],并对 4 名 GC 患者的腹水进行了单细胞 RNA 测序(scRNA-seq)分析(补充表 S3-S4)。分析流程见补充图 S2。通过对 66 个配对的原发性和转移性肿瘤进行差异表达分析,我们确定了 949 个与转移性 GC 肿瘤独特相关的基因(补充图 S3)。随后对这些转移性 GC 特异性基因的过度表达分析表明,血源性转移性肿瘤表现出受体酪氨酸激酶相关信号的上调,同时表现出上皮-间质转化(EMT)相关通路的下调(补充图 S4)。相反,腹膜/卵巢转移性肿瘤则表现出刺猬(Hedgehog)通路的上调,而刺猬通路是 EMT 信号转导的关键参与者。这些研究结果表明,GC 转移的机制因途径而异[5],因此强调了临床上需要考虑特定途径。非负因式分解(NMF)算法根据GC特异性基因的表达将TCGA队列分为两个亚型,分别称为NMF-G1和NMF-G2。NMF-G2 亚型的特点是 GC 特异性基因表达上调,与基因组稳定亚型相关。该亚型经常出现粘连蛋白 1(CDH1)的体细胞突变,而且无论哪种分子亚型,其预后都较差(补充图 S5)。通过加权基因共表达网络分析,我们发现了一个由 122 个基因组成的模块,这些基因在 NMF-G1 和 NMF-G2 之间具有不同的表达模式。值得注意的是,这些基因表现出 EMT 相关信号的上调,因此被称为转移特异性 EMT(msEMT)基因(附图 S6)。这些 msEMT 基因是独特的,因为它们不同于 GC 生物学中的既定基因集和共识癌症驱动基因 [6,9]。此外,大多数 msEMT 基因在血行转移中表现出下调。为了验证msEMT基因在GC中的临床意义,我们分析了ACRG[6]和YCC[7]两个独立的原发性GC队列中这些基因的表达水平,这两个队列都有复发数据。通过对 msEMT 基因表达谱的评估,我们将每个队列分为两个不同的亚型,其中高表达组被指定为 msEMT 亚型。重要的是,在两个队列中,即使调整了临床因素,msEMT 亚型也与明显较差的预后相关(补充图 S7A-D)。此外,原发性肿瘤中的 msEMT 亚型与腹膜/卵巢复发的关系更为密切。机器学习分析表明,msEMT 基因对腹膜/卵巢转移有很强的预测能力,准确率很高(&gt;0.
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
Deciphering metastatic route-specific signals and their microenvironment interactions in peritoneal metastasis of gastric cancer

Gastric cancer (GC) is a pervasive global malignancy with high mortality rates due to distant metastasis [1]. GC metastasis can occur via hematogenous route, peritoneal route, and specifically through ovarian spread in females [2]. Among these, peritoneal metastasis is the most prevalent and challenging condition to treat [3]. The Cancer Genome Atlas (TCGA) has uncovered four molecular subtypes of GC: microsatellite instability, Epstein-Barr virus-related, chromosomal instability, and genomically stable [4]. These subtypes exhibit unique features and metastatic behaviors [5], providing valuable insights into the molecular characteristics of GC. Nevertheless, there remains a gap in understanding gene expression feature in metastatic GC compared to corresponding primary tumors, particularly regarding its biological and clinical relevance in different metastatic patterns.

A whole-transcriptome sequencing analysis was conducted to investigate the characteristics of metastatic GC. We examined 66 paired primary and metastatic tumors, categorized according to their sites (38 primary, 9 hematogenous, 6 peritoneal, and 13 ovarian metastases), collected from 14 patients with GC metastasis (Supplementary Tables S1-S2 and Supplementary Figure S1). To corroborate the biological and clinical relevance of the identified metastatic GC-specific signals, we utilized publicly available databases containing expression data on primary GC, including TCGA (n = 415) [4], Asian Cancer Research Group (ACRG, n = 300) [6], and Yonsei Cancer Center (YCC, n = 433) cohorts [7]. For in-depth analysis at the single-cell resolution, we accessed a public dataset comprising 5 primary GC samples [8] and performed a single-cell RNA sequencing (scRNA-seq) analysis on ascites obtained from 4 patients with GC (Supplementary Tables S3-S4). The flow of analysis is summarized in Supplementary Figure S2. Details of the patient demographics, samples, and data analysis methods used are outlined in the Supplementary Methods.

Through a differential expression analysis of the 66 paired primary and metastatic tumors, we identified 949 genes uniquely associated with metastatic GC tumors (Supplementary Figure S3). Subsequent over-representation analysis of these metastatic GC-specific genes indicated that hematogenous metastatic tumors exhibited an up-regulation of receptor tyrosine kinase-related signals, while simultaneously showing a down-regulation of pathways associated with epithelial-mesenchymal transition (EMT) (Supplementary Figure S4). Conversely, peritoneal/ovarian metastatic tumors demonstrated an upregulation of the Hedgehog pathway, a key player in EMT signaling. These findings suggest that the mechanisms underlying GC metastasis vary by route [5], thus underscoring the need for route-specific clinical considerations.

To assess the clinical and biological significance of the identified genes, we examined their expression profiles in the TCGA cohort, which consisted of 415 primary GC tumors [4]. The non-negative factorization (NMF) algorithm stratified TCGA cohort into two subtypes based on the expression of GC-specific genes, denoted as NMF-G1 and NMF-G2. The NMF-G2 subtype, characterized by upregulated expression of GC-specific genes, was associated with the genomically stable subtype. It frequently exhibited somatic mutations in the cadherin 1 (CDH1) and was associated with a poor prognosis, regardless of the molecular subtypes (Supplementary Figure S5). Through weighted gene co-expression network analysis, we identified a module comprising 122 genes with distinctive expression patterns between NMF-G1 and NMF-G2. Notably, these genes exhibited the upregulation of EMT-related signals, leading to their designation as metastasis-specific EMT (msEMT) genes (Supplementary Figure S6). These msEMT genes are unique as they differ from established gene sets in GC biology and consensus cancer driver genes [6, 9]. Furthermore, the majority of msEMT genes exhibited downregulation in hematogenous metastasis. Upon re-evaluation, the expression level of these genes was higher in peritoneal/ovarian metastatic tumors than in corresponding primary tumors (Supplementary Tables S5-S6).

To validate the clinical relevance of msEMT genes in GC, we analyzed their expression levels in two independent primary GC cohorts from ACRG [6] and YCC [7], which had recurrence data. Assessment of the msEMT gene expression profile enabled the classification of each cohort into two distinct subtypes, with the high-expression group designated as the msEMT subtype. Importantly, the msEMT subtype was associated with a significantly worse prognosis in both cohorts, even after adjusting for clinical factors (Supplementary Figure S7A-D). Furthermore, the msEMT subtype in primary tumors was more closely linked to peritoneal/ovarian recurrence. Machine learning analyses showed that msEMT genes had a strong predictive capacity for peritoneal/ovarian metastasis, with high accuracy (>0.80), particularly for negative predictive values and specificity, compared with models involving clinical variables (Supplementary Figure S7E and Supplementary Table S7). These results highlight the pivotal role of msEMT genes in the occurrence of peritoneal/ovarian metastasis and suggest that primary GC possesses metastatic potential even before the onset of peritoneal/ovarian metastasis; therefore, msEMT genes could serve as potential biomarkers for risk assessment.

To understand the origin of msEMT gene expression in GC, we conducted a scRNA-seq analysis of primary tumors (stratified by superficial and deep layer) [8] and ascites from 4 patients with gastric cancer metastasis. msEMT genes were not predominantly expressed in cancer cells, but rather in fibroblasts within the deep layers of primary tumors and in ascites (Supplementary Figure S8). This result indicated that msEMT signals are likely to arise from the tumor microenvironment, particularly from cancer-associated fibroblasts (CAFs), during the late stages of primary tumor development and peritoneal metastasis.

To further understand the expression patterns of msEMT genes in specific fibroblast populations, we explored their landscape and dynamic changes in fibroblasts. Within the deep layer of primary tumors, fibroblast lineage trajectory inference revealed that classical myofibroblastic CAFs (myo-CAFs, C1) and inflammatory CAFs (i-CAFs, C0) initiated the pseudo-time trajectory, whereas CAFs associated with cancer migration and EMT (C2) were at the end of the trajectory. msEMT genes exhibited differential expression across these fibroblast subtypes (Supplementary Figure S9A). In the ascites, vascular and inflammatory CAFs (C1 and C3) initiated the pseudo-time trajectory, myo-CAFs (C5) were positioned in the middle of the trajectory, and the mature CAFs associated with CAF proliferation and cancer cell migration (C6 and C7) concluded the trajectory (Supplementary Figure S9B). msEMT genes were mainly expressed across specific fibroblast subtypes, excepts myo-CAFs (C5). This implies their involvement in modulating the cellular landscape of fibroblasts during GC peritoneal metastasis.

When the gene expression profiles of fibroblasts within primary tumors were compared with those in ascites, we observed distinctly upregulated signals of extracellular matrix modulation in primary tumors, whereas ascites exhibited the upregulation of signals associated with cell-cell adhesion and immune response (Supplementary Figure S10). The interactions between fibroblasts and other cellular components are enhanced in ascites than in primary tumors, emphasizing the importance of comprehending cell-to-cell interactions in these contexts.

Cell-to-cell interaction analysis revealed that fibroblasts orchestrated and responded to signals associated with tumor growth and progression in primary tumors (Supplementary Figure S11A). Conversely, the fibroblasts within ascites predominantly receive signals associated with the modulation of angiogenesis and immune evasion, such as tumor necrosis factor-related weak inducer of apoptosis, complement, and macrophage migration inhibitory factor (Supplementary Figure S11B). The changes in fibroblast signaling reflect the alterations in the dynamic interplay between cancer cells and fibroblasts throughout the metastatic cascade, highlighting their pivotal role in the peritoneal dissemination of GC.

Our study has delineated the metastatic route-specific molecular profiles of GC, highlighting the prognostic impact of msEMT genes and the crucial role of CAFs in GC progression and peritoneal spread (Figure 1). These insights enhance our understanding of GC molecular variations by metastatic route and underscore the contribution of the tumor microenvironment to the dissemination of GC.

This study was approved by the Institutional Review Board of Severance Hospital (4-2019-0188) and Soonchunhyang University Bucheon Hospital (2022-09-008), and informed consent was received from all patients.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Cancer Communications
Cancer Communications Biochemistry, Genetics and Molecular Biology-Cancer Research
CiteScore
25.50
自引率
4.30%
发文量
153
审稿时长
4 weeks
期刊介绍: Cancer Communications is an open access, peer-reviewed online journal that encompasses basic, clinical, and translational cancer research. The journal welcomes submissions concerning clinical trials, epidemiology, molecular and cellular biology, and genetics.
期刊最新文献
Advances in cervical cancer: current insights and future directions. PTPN9 regulates HER3 phosphorylation during trastuzumab treatment and loss of PTPN9 is a potential biomarker for trastuzumab resistance in HER2 positive breast cancer. Anti-tumor drug supervision in China from 2010 to 2024: the evolution and prospect of drug review standards. Ex vivo STAT3 phosphorylation in circulating immune cells: a novel biomarker for early cancer diagnosis and response to anti-PD-1 therapy. Issue Information
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1