首页 > 最新文献

Molecular Cancer最新文献

英文 中文
A case of response to combination treatment with TSA-DC-CTL immunotherapy and osimertinib in EGFR mutated advanced lung adenocarcinoma 一例表皮生长因子受体突变晚期肺腺癌患者对TSA-DC-CTL免疫疗法和奥希替尼联合治疗的应答病例
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-09 DOI: 10.1186/s12943-024-02070-3
Zhiyi Han, Tao Li, Heng Zhang, Kai Liang, Mingcong You, Mengdi Xu, Fan Bai, Tongmei Zhang
This study details a case of a patient with advanced lung adenocarcinoma harboring an exon 19 deletion in the EGFR gene. A 46-year-old female patient was diagnosed with stage IVb left lung adenocarcinoma, with multiple bone and lymph node metastases. Following the identification of tumor-specific antigen peptides, the patient received a combination treatment of immunotherapy (TSA-DC-CTL) and oral osimertinib. Peripheral blood circulating immune cells and circulating tumor cells (CTCs) were monitored before and after treatment. PET-CT and CT scans were used to assess the tumor response to treatment. A significant increase in total lymphocyte percentage and decrease in the number of CTCs in the patient was observed. Imaging studies showed a notable reduction in tumor metastases. This report demonstrates the safety and efficacy of TSA-DC-CTL cell immunotherapy combined with osimertinib in the treatment of a patient with advanced lung adenocarcinoma with an EGFR exon 19 deletions. This study describes a promising new treatment option for patients with advanced lung cancer with EGFR mutations.
本研究详细介绍了一例表皮生长因子受体基因第19外显子缺失的晚期肺腺癌患者。一名 46 岁的女性患者被诊断为左肺腺癌 IVb 期,并伴有多处骨和淋巴结转移。在鉴定出肿瘤特异性抗原肽后,患者接受了免疫疗法(TSA-DC-CTL)和口服奥希替尼的联合治疗。治疗前后对外周血循环免疫细胞和循环肿瘤细胞(CTC)进行了监测。PET-CT 和 CT 扫描用于评估肿瘤对治疗的反应。结果显示,患者的总淋巴细胞比例明显增加,CTC数量明显减少。影像学研究显示肿瘤转移明显减少。本报告展示了 TSA-DC-CTL 细胞免疫疗法联合奥希替尼治疗表皮生长因子受体外显子 19 缺失的晚期肺腺癌患者的安全性和有效性。这项研究为表皮生长因子受体(EGFR)突变的晚期肺癌患者提供了一种前景广阔的新治疗方案。
{"title":"A case of response to combination treatment with TSA-DC-CTL immunotherapy and osimertinib in EGFR mutated advanced lung adenocarcinoma","authors":"Zhiyi Han, Tao Li, Heng Zhang, Kai Liang, Mingcong You, Mengdi Xu, Fan Bai, Tongmei Zhang","doi":"10.1186/s12943-024-02070-3","DOIUrl":"https://doi.org/10.1186/s12943-024-02070-3","url":null,"abstract":"This study details a case of a patient with advanced lung adenocarcinoma harboring an exon 19 deletion in the EGFR gene. A 46-year-old female patient was diagnosed with stage IVb left lung adenocarcinoma, with multiple bone and lymph node metastases. Following the identification of tumor-specific antigen peptides, the patient received a combination treatment of immunotherapy (TSA-DC-CTL) and oral osimertinib. Peripheral blood circulating immune cells and circulating tumor cells (CTCs) were monitored before and after treatment. PET-CT and CT scans were used to assess the tumor response to treatment. A significant increase in total lymphocyte percentage and decrease in the number of CTCs in the patient was observed. Imaging studies showed a notable reduction in tumor metastases. This report demonstrates the safety and efficacy of TSA-DC-CTL cell immunotherapy combined with osimertinib in the treatment of a patient with advanced lung adenocarcinoma with an EGFR exon 19 deletions. This study describes a promising new treatment option for patients with advanced lung cancer with EGFR mutations.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141908943","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Efficacy and safety of chiauranib in a combination therapy in platinum-resistant or refractory ovarian cancer: a multicenter, open-label, phase Ib and II study 巧拉尼联合疗法治疗铂类耐药或难治性卵巢癌的疗效和安全性:一项多中心、开放标签、Ib 期和 II 期研究
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-09 DOI: 10.1186/s12943-024-02076-x
Jin Li, Jihong Liu, Rutie Yin, Dongling Zou, Hong Zheng, Junning Cao, Zhendong Chen, Wei Sun, Yunong Gao, Songling Zhang, Linjuan Zeng, Ruifang An, Xianping Lu, Shuang Ye, Xiaohua Wu
Platinum-resistant or refractory ovarian cancer is a highly lethal gynecologic disease with limited treatment options. Chiauranib is a novel small-molecule selective inhibitor, which could effectively target multiple pathways including Aurora B and CSF-1R to inhibit cell cycle process and improve anti-tumor immune function, as long as VEGF pathway for tumor extinction. A phase II study was sequentially conducted after a phase Ib monotherapy study to evaluate the efficacy of chiauranib combined with chemotherapy. Chinese patients with recurrent ovarian cancer were enrolled. Eligible patients received chiauranib combined with a maximum of six cycles of chemotherapy: etoposide (CE group) or weekly-paclitaxel (CP group). Patients, who exhibited a complete or partial response, or stable disease following combo treatment, progressed to maintenance phase to receive chiauranib monotherapy. Primary endpoint was progression-free survival (PFS) according to RECIST v1.1. From November 2017 to March 2019, 25 patients were enrolled in a phase 1b study and a median PFS of 3.7 months (95% CI 1.8–NE) was achieved by chiauranib monotherapy. From July 2019 to December 2020, a total of 47 patients were enrolled in the phase II study. One CP patient did not receive the study drugs, and three patients withdrew before the first tumor assessment. Thus, 43 patients (CE group: 22 patients; CP group: 21 patients) were included in the evaluation. The median PFS was 5·4 months (95% CI 2·8–5·6) and 5·6 months (95% CI 3·4–7·0), respectively. This was the first study to evaluate chiauranib, a novel multi-targeted kinase inhibitor in patients with ovarian cancer. The administration of chiauranib along with etoposide or weekly-paclitaxel significantly enhanced the efficacy with manageable adverse events. This warrants further clinical studies on this novel treatment. A phase III study is promising and ongoing. ClinicaTrials.gov identifier: NCT03901118 (phase II) and NCT03166891 (phase Ib).
铂耐药或难治性卵巢癌是一种致死率极高的妇科疾病,但治疗方案却十分有限。Chiauranib是一种新型小分子选择性抑制剂,可有效靶向包括Aurora B和CSF-1R在内的多条通路,抑制细胞周期过程,提高抗肿瘤免疫功能,同时也可靶向VEGF通路消灭肿瘤。在Ib期单药治疗研究之后,我们又进行了一项II期研究,以评估chiauranib联合化疗的疗效。研究招募了中国复发性卵巢癌患者。符合条件的患者接受巧拉尼联合化疗,最多6个周期:依托泊苷(CE组)或每周紫杉醇(CP组)。如果患者在联合治疗后出现完全或部分应答,或病情稳定,则进入维持治疗阶段,接受chiauranib单药治疗。根据RECIST v1.1标准,主要终点为无进展生存期(PFS)。2017年11月至2019年3月,25名患者入组1b期研究,接受chiauranib单药治疗的中位PFS为3.7个月(95% CI 1.8-NE)。2019年7月至2020年12月,共有47名患者入组II期研究。一名 CP 患者未接受研究药物,三名患者在首次肿瘤评估前退出。因此,43 名患者(CE 组:22 名患者;CP 组:21 名患者)被纳入评估。中位生存期分别为 5-4 个月(95% CI 2-8-5-6 )和 5-6 个月(95% CI 3-4-7-0)。这是第一项评估卵巢癌患者使用新型多靶点激酶抑制剂chiauranib的研究。chiauranib与依托泊苷或每周紫杉醇合用可显著提高疗效,且不良反应可控。因此,有必要对这种新型疗法进行进一步的临床研究。目前正在进行的III期研究前景广阔。ClinicaTrials.gov 标识符:NCT03901118(II 期)和 NCT03166891(Ib 期)。
{"title":"Efficacy and safety of chiauranib in a combination therapy in platinum-resistant or refractory ovarian cancer: a multicenter, open-label, phase Ib and II study","authors":"Jin Li, Jihong Liu, Rutie Yin, Dongling Zou, Hong Zheng, Junning Cao, Zhendong Chen, Wei Sun, Yunong Gao, Songling Zhang, Linjuan Zeng, Ruifang An, Xianping Lu, Shuang Ye, Xiaohua Wu","doi":"10.1186/s12943-024-02076-x","DOIUrl":"https://doi.org/10.1186/s12943-024-02076-x","url":null,"abstract":"Platinum-resistant or refractory ovarian cancer is a highly lethal gynecologic disease with limited treatment options. Chiauranib is a novel small-molecule selective inhibitor, which could effectively target multiple pathways including Aurora B and CSF-1R to inhibit cell cycle process and improve anti-tumor immune function, as long as VEGF pathway for tumor extinction. A phase II study was sequentially conducted after a phase Ib monotherapy study to evaluate the efficacy of chiauranib combined with chemotherapy. Chinese patients with recurrent ovarian cancer were enrolled. Eligible patients received chiauranib combined with a maximum of six cycles of chemotherapy: etoposide (CE group) or weekly-paclitaxel (CP group). Patients, who exhibited a complete or partial response, or stable disease following combo treatment, progressed to maintenance phase to receive chiauranib monotherapy. Primary endpoint was progression-free survival (PFS) according to RECIST v1.1. From November 2017 to March 2019, 25 patients were enrolled in a phase 1b study and a median PFS of 3.7 months (95% CI 1.8–NE) was achieved by chiauranib monotherapy. From July 2019 to December 2020, a total of 47 patients were enrolled in the phase II study. One CP patient did not receive the study drugs, and three patients withdrew before the first tumor assessment. Thus, 43 patients (CE group: 22 patients; CP group: 21 patients) were included in the evaluation. The median PFS was 5·4 months (95% CI 2·8–5·6) and 5·6 months (95% CI 3·4–7·0), respectively. This was the first study to evaluate chiauranib, a novel multi-targeted kinase inhibitor in patients with ovarian cancer. The administration of chiauranib along with etoposide or weekly-paclitaxel significantly enhanced the efficacy with manageable adverse events. This warrants further clinical studies on this novel treatment. A phase III study is promising and ongoing. ClinicaTrials.gov identifier: NCT03901118 (phase II) and NCT03166891 (phase Ib).","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141908944","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Refining the diagnostic utility of OLFM4 in gastric cancer precursors: a call for rigorous methodologies 完善 OLFM4 在胃癌前体中的诊断作用:呼吁采用严格的方法学
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-08 DOI: 10.1186/s12943-024-02077-w
Tai Zhang, Xudong Tang
This commentary offers a thoughtful discussion of the study by Wei et al. published in the journal on the role of Olfactomedin 4 (OLFM4) in incomplete intestinal metaplasia, a gastric precancerous condition. The original paper introduces OLFM4 as a novel biomarker with potential enhanced diagnostic efficacy compared to established markers. However, several methodological and interpretive considerations are noted. The histopathological findings could be refined by using higher magnification to better elucidate the cellular localization of OLFM4. Including high-resolution images for key stainings would enhance the study’s robustness in expression profiling. The statistical approach could be strengthened by employing more rigorous, quantitative methodologies. Additionally, integrating immunofluorescence double-staining may improve the reliability of the results. Discrepancies in immunohistochemical signals across datasets suggest a need for further investigation into tissue section representativeness. Clarifying the term “precancerous lesions of gastric carcinoma cells” to align with widely accepted definitions would enhance clarity. The choice of the GES-1 cell model treated with MNNG could be reconsidered in favor of more established models such as organoids, air-liquid interface models, and gastric cancer-specific cell lines. The in vivo MNNG-alcohol combination model might require additional empirical support, given the limited and conflicting literature on this approach, to ensure an accurate portrayal of IM pathogenesis. The commentary concludes with a call for stringent and standardized methodologies in biomarker research to ensure the clinical applicability and reliability of biomarker studies, particularly in the context of gastric cancer detection and intervention.
这篇评论对 Wei 等人发表在该杂志上的研究进行了深思熟虑的讨论,该研究涉及 Olfactomedin 4 (OLFM4) 在胃癌前病变--不完全性肠化生中的作用。原论文将 OLFM4 介绍为一种新型生物标记物,与已有的标记物相比,其诊断效果可能更强。不过,本文也指出了一些方法学和解释学方面的注意事项。组织病理学研究结果可以通过使用更高的放大倍率来完善,以更好地阐明 OLFM4 的细胞定位。将关键染色的高分辨率图像纳入研究将提高表达谱分析的稳健性。统计方法可以通过采用更严格的定量方法来加强。此外,整合免疫荧光双重染色可提高结果的可靠性。不同数据集之间免疫组化信号的差异表明,有必要进一步调查组织切片的代表性。明确 "胃癌细胞癌前病变 "这一术语,使其与广泛接受的定义相一致,将使结果更加清晰。可以重新考虑选择用 MNNG 处理的 GES-1 细胞模型,转而使用器官组织、气液界面模型和胃癌特异性细胞系等更成熟的模型。鉴于有关这种方法的文献有限且相互矛盾,体内 MNNG-酒精组合模型可能需要更多的经验支持,以确保准确描述 IM 的发病机制。评论最后呼吁在生物标志物研究中采用严格的标准化方法,以确保生物标志物研究的临床适用性和可靠性,尤其是在胃癌检测和干预方面。
{"title":"Refining the diagnostic utility of OLFM4 in gastric cancer precursors: a call for rigorous methodologies","authors":"Tai Zhang, Xudong Tang","doi":"10.1186/s12943-024-02077-w","DOIUrl":"https://doi.org/10.1186/s12943-024-02077-w","url":null,"abstract":"This commentary offers a thoughtful discussion of the study by Wei et al. published in the journal on the role of Olfactomedin 4 (OLFM4) in incomplete intestinal metaplasia, a gastric precancerous condition. The original paper introduces OLFM4 as a novel biomarker with potential enhanced diagnostic efficacy compared to established markers. However, several methodological and interpretive considerations are noted. The histopathological findings could be refined by using higher magnification to better elucidate the cellular localization of OLFM4. Including high-resolution images for key stainings would enhance the study’s robustness in expression profiling. The statistical approach could be strengthened by employing more rigorous, quantitative methodologies. Additionally, integrating immunofluorescence double-staining may improve the reliability of the results. Discrepancies in immunohistochemical signals across datasets suggest a need for further investigation into tissue section representativeness. Clarifying the term “precancerous lesions of gastric carcinoma cells” to align with widely accepted definitions would enhance clarity. The choice of the GES-1 cell model treated with MNNG could be reconsidered in favor of more established models such as organoids, air-liquid interface models, and gastric cancer-specific cell lines. The in vivo MNNG-alcohol combination model might require additional empirical support, given the limited and conflicting literature on this approach, to ensure an accurate portrayal of IM pathogenesis. The commentary concludes with a call for stringent and standardized methodologies in biomarker research to ensure the clinical applicability and reliability of biomarker studies, particularly in the context of gastric cancer detection and intervention.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141904590","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Essential gene screening identifies the bromodomain-containing protein BRPF1 as a new actionable target for endocrine therapy-resistant breast cancers 基本基因筛选确定含溴结构域蛋白 BRPF1 为内分泌治疗耐药乳腺癌的新靶点
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-07 DOI: 10.1186/s12943-024-02071-2
Annamaria Salvati, Giorgio Giurato, Jessica Lamberti, Ilaria Terenzi, Laura Crescenzo, Viola Melone, Luigi Palo, Alessandro Giordano, Francesco Sabbatino, Giuseppina Roscigno, Cristina Quintavalle, Gerolama Condorelli, Francesca Rizzo, Roberta Tarallo, Giovanni Nassa, Alessandro Weisz
Identifying master epigenetic factors controlling proliferation and survival of cancer cells allows to discover new molecular targets exploitable to overcome resistance to current pharmacological regimens. In breast cancer (BC), resistance to endocrine therapy (ET) arises from aberrant Estrogen Receptor alpha (ERα) signaling caused by genetic and epigenetic events still mainly unknown. Targeting key upstream components of the ERα pathway provides a way to interfere with estrogen signaling in cancer cells independently from any other downstream event. By combining computational analysis of genome-wide ‘drop-out’ screenings with siRNA-mediated gene knock-down (kd), we identified a set of essential genes in luminal-like, ERα + BC that includes BRPF1, encoding a bromodomain-containing protein belonging to a family of epigenetic readers that act as chromatin remodelers to control gene transcription. To gather mechanistic insights into the role of BRPF1 in BC and ERα signaling, we applied chromatin and transcriptome profiling, gene ablation and targeted pharmacological inhibition coupled to cellular and functional assays. Results indicate that BRPF1 associates with ERα onto BC cell chromatin and its blockade inhibits cell cycle progression, reduces cell proliferation and mediates transcriptome changes through the modulation of chromatin accessibility. This effect is elicited by a widespread inhibition of estrogen signaling, consequent to ERα gene silencing, in antiestrogen (AE) -sensitive and -resistant BC cells and pre-clinical patient-derived models (PDOs). Characterization of the functional interplay of BRPF1 with ERα reveals a new regulator of estrogen-responsive BC cell survival and suggests that this epigenetic factor is a potential new target for treatment of these tumors.
确定控制癌细胞增殖和存活的主表观遗传因素,有助于发现新的分子靶点,从而克服对现有药物疗法的耐药性。在乳腺癌(BC)中,对内分泌疗法(ET)的耐药性源于雌激素受体α(ERα)信号传导异常,其原因主要是遗传和表观遗传事件,目前仍不清楚。靶向雌激素受体α通路的关键上游成分为干扰癌细胞中的雌激素信号提供了一种独立于任何其他下游事件的方法。通过将全基因组 "退出 "筛选的计算分析与 siRNA 介导的基因敲除 (kd) 相结合,我们确定了一组管腔样、ERα + BC 中的重要基因,其中包括 BRPF1,它编码一种含溴结构域的蛋白,属于表观遗传阅读器家族,可作为染色质重塑器控制基因转录。为了从机理上深入了解BRPF1在BC和ERα信号转导中的作用,我们应用了染色质和转录组图谱分析、基因消减和靶向药理抑制以及细胞和功能测试。结果表明,BRPF1与ERα在BC细胞染色质上结合,阻断BRPF1可抑制细胞周期的进展,减少细胞增殖,并通过调节染色质的可及性介导转录组的变化。在抗雌激素(AE)敏感和耐受的 BC 细胞以及临床前患者衍生模型(PDOs)中,ERα 基因沉默导致的雌激素信号传导受到广泛抑制,从而产生了这种效应。BRPF1与ERα功能相互作用的特征揭示了雌激素反应性BC细胞存活的新调节因子,并表明这种表观遗传因子是治疗这些肿瘤的潜在新靶点。
{"title":"Essential gene screening identifies the bromodomain-containing protein BRPF1 as a new actionable target for endocrine therapy-resistant breast cancers","authors":"Annamaria Salvati, Giorgio Giurato, Jessica Lamberti, Ilaria Terenzi, Laura Crescenzo, Viola Melone, Luigi Palo, Alessandro Giordano, Francesco Sabbatino, Giuseppina Roscigno, Cristina Quintavalle, Gerolama Condorelli, Francesca Rizzo, Roberta Tarallo, Giovanni Nassa, Alessandro Weisz","doi":"10.1186/s12943-024-02071-2","DOIUrl":"https://doi.org/10.1186/s12943-024-02071-2","url":null,"abstract":"Identifying master epigenetic factors controlling proliferation and survival of cancer cells allows to discover new molecular targets exploitable to overcome resistance to current pharmacological regimens. In breast cancer (BC), resistance to endocrine therapy (ET) arises from aberrant Estrogen Receptor alpha (ERα) signaling caused by genetic and epigenetic events still mainly unknown. Targeting key upstream components of the ERα pathway provides a way to interfere with estrogen signaling in cancer cells independently from any other downstream event. By combining computational analysis of genome-wide ‘drop-out’ screenings with siRNA-mediated gene knock-down (kd), we identified a set of essential genes in luminal-like, ERα + BC that includes BRPF1, encoding a bromodomain-containing protein belonging to a family of epigenetic readers that act as chromatin remodelers to control gene transcription. To gather mechanistic insights into the role of BRPF1 in BC and ERα signaling, we applied chromatin and transcriptome profiling, gene ablation and targeted pharmacological inhibition coupled to cellular and functional assays. Results indicate that BRPF1 associates with ERα onto BC cell chromatin and its blockade inhibits cell cycle progression, reduces cell proliferation and mediates transcriptome changes through the modulation of chromatin accessibility. This effect is elicited by a widespread inhibition of estrogen signaling, consequent to ERα gene silencing, in antiestrogen (AE) -sensitive and -resistant BC cells and pre-clinical patient-derived models (PDOs). Characterization of the functional interplay of BRPF1 with ERα reveals a new regulator of estrogen-responsive BC cell survival and suggests that this epigenetic factor is a potential new target for treatment of these tumors.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141899444","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Circular RNA ACVR2A promotes the progression of hepatocellular carcinoma through mir-511-5p targeting PI3K-Akt signaling pathway 环状 RNA ACVR2A 通过 mir-511-5p 靶向 PI3K-Akt 信号通路促进肝细胞癌的进展
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-06 DOI: 10.1186/s12943-024-02074-z
Du Fei, Fang Wang, Yaohui Wang, Ji Chen, Shendong Chen, Lianpeng Fan, Luhan Yang, Qingyi Ren, Suwit Duangmano, Fukuan Du, Hao Liu, Jie Zhou, Jing Sheng, Yueshui Zhao, Xu Wu, Mingxing Li, Zhangang Xiao, Zhuo Zhang, Xian Jiang
Circular RNA (circRNA) is thought to mediate the occurrence and development of human cancer and usually acts as a tiny RNA (miRNA) sponge to regulate downstream gene expression. However, it is not clear whether and how circACVR2A (hsa_circ_0001073) is involved in the progression of HCC. The purpose of this study is to clarify the potential role and molecular mechanism of circACVR2A in regulating the progression of hepatocellular carcinoma cells (HCC). The abundance of related proteins in circACVR2A, microRNA (miR511-5p) and PI3K-Akt signaling pathway was determined by quantitative reverse transcriptase polymerase chain reaction (RT-PCR) or Western blotting. Cell viability, invasion and apoptosis were analyzed by CCK-8, Transwell analysis and Tunel staining, respectively. The interaction between circACVR2A and microRNA was evaluated by double luciferase reporter gene assay. The results showed that circACVR2A was highly expressed in hepatocellular carcinoma cell lines. Our in vivo and in vitro data showed that circACVR2A promoted the proliferation, migration and invasion of HCC. In terms of mechanism, we found that circACVR2A can directly interact with miR511-5p and act as a miRNA sponge to regulate the expression of related proteins in PI3K-Akt signaling pathway. In HCC, circACVR2A can mediate miR-511-5p/mRNA network to activate PI3K signal pathway. This shows that the molecular regulatory network with circACVR2A as the core is a new potential target for diagnosis and treatment of hepatocellular carcinoma.
环状 RNA(circRNA)被认为介导人类癌症的发生和发展,通常作为微小 RNA(miRNA)海绵调控下游基因的表达。然而,尚不清楚 circACVR2A (hsa_circ_0001073) 是否以及如何参与 HCC 的进展。本研究旨在阐明 circACVR2A 在调控肝癌细胞(HCC)进展中的潜在作用和分子机制。研究采用定量逆转录酶聚合酶链反应(RT-PCR)或 Western 印迹法测定了 circACVR2A、microRNA(miR511-5p)和 PI3K-Akt 信号通路中相关蛋白的丰度。细胞活力、侵袭和凋亡分别通过 CCK-8、Transwell 分析和 Tunel 染色进行分析。通过双荧光素酶报告基因实验评估了 circACVR2A 与 microRNA 之间的相互作用。结果显示,circACVR2A 在肝癌细胞系中高表达。我们的体内和体外数据显示,circACVR2A 促进了 HCC 的增殖、迁移和侵袭。在机制方面,我们发现 circACVR2A 可直接与 miR511-5p 相互作用,并作为 miRNA 海绵调控 PI3K-Akt 信号通路中相关蛋白的表达。在 HCC 中,circACVR2A 可介导 miR-511-5p/mRNA 网络激活 PI3K 信号通路。这表明,以 circACVR2A 为核心的分子调控网络是诊断和治疗肝细胞癌的潜在新靶点。
{"title":"Circular RNA ACVR2A promotes the progression of hepatocellular carcinoma through mir-511-5p targeting PI3K-Akt signaling pathway","authors":"Du Fei, Fang Wang, Yaohui Wang, Ji Chen, Shendong Chen, Lianpeng Fan, Luhan Yang, Qingyi Ren, Suwit Duangmano, Fukuan Du, Hao Liu, Jie Zhou, Jing Sheng, Yueshui Zhao, Xu Wu, Mingxing Li, Zhangang Xiao, Zhuo Zhang, Xian Jiang","doi":"10.1186/s12943-024-02074-z","DOIUrl":"https://doi.org/10.1186/s12943-024-02074-z","url":null,"abstract":"Circular RNA (circRNA) is thought to mediate the occurrence and development of human cancer and usually acts as a tiny RNA (miRNA) sponge to regulate downstream gene expression. However, it is not clear whether and how circACVR2A (hsa_circ_0001073) is involved in the progression of HCC. The purpose of this study is to clarify the potential role and molecular mechanism of circACVR2A in regulating the progression of hepatocellular carcinoma cells (HCC). The abundance of related proteins in circACVR2A, microRNA (miR511-5p) and PI3K-Akt signaling pathway was determined by quantitative reverse transcriptase polymerase chain reaction (RT-PCR) or Western blotting. Cell viability, invasion and apoptosis were analyzed by CCK-8, Transwell analysis and Tunel staining, respectively. The interaction between circACVR2A and microRNA was evaluated by double luciferase reporter gene assay. The results showed that circACVR2A was highly expressed in hepatocellular carcinoma cell lines. Our in vivo and in vitro data showed that circACVR2A promoted the proliferation, migration and invasion of HCC. In terms of mechanism, we found that circACVR2A can directly interact with miR511-5p and act as a miRNA sponge to regulate the expression of related proteins in PI3K-Akt signaling pathway. In HCC, circACVR2A can mediate miR-511-5p/mRNA network to activate PI3K signal pathway. This shows that the molecular regulatory network with circACVR2A as the core is a new potential target for diagnosis and treatment of hepatocellular carcinoma.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141895346","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
BRCA1 secondary splice-site mutations drive exon-skipping and PARP inhibitor resistance. BRCA1 次级剪接位点突变驱动外显子跳转和 PARP 抑制剂抗性。
IF 27.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-05 DOI: 10.1186/s12943-024-02048-1
Ksenija Nesic, John J Krais, Yifan Wang, Cassandra J Vandenberg, Pooja Patel, Kathy Q Cai, Tanya Kwan, Elizabeth Lieschke, Gwo-Yaw Ho, Holly E Barker, Justin Bedo, Silvia Casadei, Andrew Farrell, Marc Radke, Kristy Shield-Artin, Jocelyn S Penington, Franziska Geissler, Elizabeth Kyran, Robert Betsch, Lijun Xu, Fan Zhang, Alexander Dobrovic, Inger Olesen, Rebecca Kristeleit, Amit Oza, Iain McNeish, Gayanie Ratnayake, Nadia Traficante, Anna DeFazio, David D L Bowtell, Thomas C Harding, Kevin Lin, Elizabeth M Swisher, Olga Kondrashova, Clare L Scott, Neil Johnson, Matthew J Wakefield

PARP inhibitor (PARPi) therapy has transformed outcomes for patients with homologous recombination DNA repair (HRR) deficient ovarian cancers, for example those with BRCA1 or BRCA2 gene defects. Unfortunately, PARPi resistance is common. Multiple resistance mechanisms have been described, including secondary mutations that restore the HR gene reading frame. BRCA1 splice isoforms △11 and △11q can contribute to PARPi resistance by splicing out the mutation-containing exon, producing truncated, partially functional proteins. However, the clinical impacts and underlying drivers of BRCA1 exon skipping are not fully understood.We analyzed nine ovarian and breast cancer patient derived xenografts (PDX) with BRCA1 exon 11 frameshift mutations for exon skipping and therapy response, including a matched PDX pair derived from a patient pre- and post-chemotherapy/PARPi. BRCA1 exon 11 skipping was elevated in PARPi resistant PDX tumors. Two independent PDX models acquired secondary BRCA1 splice site mutations (SSMs) that drive exon skipping, confirmed using qRT-PCR, RNA sequencing, immunoblotting and minigene modelling. CRISPR/Cas9-mediated disruption of splicing functionally validated exon skipping as a mechanism of PARPi resistance. SSMs were also enriched in post-PARPi ovarian cancer patient cohorts from the ARIEL2 and ARIEL4 clinical trials.Few PARPi resistance mechanisms have been confirmed in the clinical setting. While secondary/reversion mutations typically restore a gene's reading frame, we have identified secondary mutations in patient cohorts that hijack splice sites to enhance mutation-containing exon skipping, resulting in the overexpression of BRCA1 hypomorphs, which in turn promote PARPi resistance. Thus, BRCA1 SSMs can and should be clinically monitored, along with frame-restoring secondary mutations.

PARP抑制剂(PARPi)疗法改变了同源重组DNA修复(HRR)缺陷卵巢癌患者(如BRCA1或BRCA2基因缺陷患者)的治疗结果。遗憾的是,PARPi 耐药性很常见。目前已描述了多种耐药机制,包括恢复 HR 基因阅读框的继发性突变。BRCA1 的剪接异构体△11 和△11q 可以剪接出含有突变的外显子,产生截短的、部分功能的蛋白质,从而导致 PARPi 耐药。我们分析了9个卵巢癌和乳腺癌患者衍生异种移植物(PDX)的BRCA1外显子11框移位突变的外显子跳过和治疗反应,包括化疗/PARPi前后患者衍生的一对匹配的PDX。在 PARPi 耐药的 PDX 肿瘤中,BRCA1 第 11 号外显子跳越率升高。利用 qRT-PCR、RNA 测序、免疫印迹和迷你基因建模证实,两个独立的 PDX 模型获得了驱动外显子跳越的继发性 BRCA1 剪接位点突变 (SSM)。CRISPR/Cas9 介导的剪接破坏功能验证了外显子跳过是 PARPi 抗性的一种机制。ARIEL2和ARIEL4临床试验的PARPi后卵巢癌患者队列中也富集了SSMs。虽然继发性/反转突变通常会恢复基因的阅读框,但我们在患者队列中发现了继发性突变,这些突变会劫持剪接位点以增强含突变外显子的跳过,从而导致 BRCA1 低形体的过度表达,进而促进 PARPi 抗性。因此,BRCA1 SSM 可以而且应该与框架恢复性继发性突变一起进行临床监测。
{"title":"BRCA1 secondary splice-site mutations drive exon-skipping and PARP inhibitor resistance.","authors":"Ksenija Nesic, John J Krais, Yifan Wang, Cassandra J Vandenberg, Pooja Patel, Kathy Q Cai, Tanya Kwan, Elizabeth Lieschke, Gwo-Yaw Ho, Holly E Barker, Justin Bedo, Silvia Casadei, Andrew Farrell, Marc Radke, Kristy Shield-Artin, Jocelyn S Penington, Franziska Geissler, Elizabeth Kyran, Robert Betsch, Lijun Xu, Fan Zhang, Alexander Dobrovic, Inger Olesen, Rebecca Kristeleit, Amit Oza, Iain McNeish, Gayanie Ratnayake, Nadia Traficante, Anna DeFazio, David D L Bowtell, Thomas C Harding, Kevin Lin, Elizabeth M Swisher, Olga Kondrashova, Clare L Scott, Neil Johnson, Matthew J Wakefield","doi":"10.1186/s12943-024-02048-1","DOIUrl":"10.1186/s12943-024-02048-1","url":null,"abstract":"<p><p>PARP inhibitor (PARPi) therapy has transformed outcomes for patients with homologous recombination DNA repair (HRR) deficient ovarian cancers, for example those with BRCA1 or BRCA2 gene defects. Unfortunately, PARPi resistance is common. Multiple resistance mechanisms have been described, including secondary mutations that restore the HR gene reading frame. BRCA1 splice isoforms △11 and △11q can contribute to PARPi resistance by splicing out the mutation-containing exon, producing truncated, partially functional proteins. However, the clinical impacts and underlying drivers of BRCA1 exon skipping are not fully understood.We analyzed nine ovarian and breast cancer patient derived xenografts (PDX) with BRCA1 exon 11 frameshift mutations for exon skipping and therapy response, including a matched PDX pair derived from a patient pre- and post-chemotherapy/PARPi. BRCA1 exon 11 skipping was elevated in PARPi resistant PDX tumors. Two independent PDX models acquired secondary BRCA1 splice site mutations (SSMs) that drive exon skipping, confirmed using qRT-PCR, RNA sequencing, immunoblotting and minigene modelling. CRISPR/Cas9-mediated disruption of splicing functionally validated exon skipping as a mechanism of PARPi resistance. SSMs were also enriched in post-PARPi ovarian cancer patient cohorts from the ARIEL2 and ARIEL4 clinical trials.Few PARPi resistance mechanisms have been confirmed in the clinical setting. While secondary/reversion mutations typically restore a gene's reading frame, we have identified secondary mutations in patient cohorts that hijack splice sites to enhance mutation-containing exon skipping, resulting in the overexpression of BRCA1 hypomorphs, which in turn promote PARPi resistance. Thus, BRCA1 SSMs can and should be clinically monitored, along with frame-restoring secondary mutations.</p>","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":27.7,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11299415/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141893866","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Genome-wide CRISPR screening identifies tyrosylprotein sulfotransferase-2 as a target for augmenting anti-PD1 efficacy 全基因组CRISPR筛选确定酪氨酸蛋白磺基转移酶-2为增强抗PD1疗效的靶点
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-02 DOI: 10.1186/s12943-024-02068-x
Yumi Oh, Sujeong Kim, Yunjae Kim, Hyun Kim, Dongjun Jang, Seungjae Shin, Soo-Jin Lee, Jiwon Kim, Sang Eun Lee, Jaeik Oh, Yoojin Yang, Dohee Kim, Hae Rim Jung, Sangjin Kim, Jihui Kim, Kyungchan Min, Beomki Cho, Hoseok Seo, Dohyun Han, Hansoo Park, Sung-Yup Cho
Immune checkpoint therapy (ICT) provides durable responses in select cancer patients, yet resistance remains a significant challenge, prompting the exploration of underlying molecular mechanisms. Tyrosylprotein sulfotransferase-2 (TPST2), known for its role in protein tyrosine O-sulfation, has been suggested to modulate the extracellular protein-protein interactions, but its specific role in cancer immunity remains largely unexplored. To explore tumor cell-intrinsic factors influencing anti-PD1 responsiveness, we conducted a pooled loss-of-function genetic screen in humanized mice engrafted with human immune cells. The responsiveness of cancer cells to interferon-γ (IFNγ) was estimated by evaluating IFNγ-mediated induction of target genes, STAT1 phosphorylation, HLA expression, and cell growth suppression. The sulfotyrosine-modified target gene of TPST2 was identified by co-immunoprecipitation and mass spectrometry. The in vivo effects of TPST2 inhibition were evaluated using mouse syngeneic tumor models and corroborated by bulk and single-cell RNA sequencing analyses. Through in vivo genome-wide CRISPR screening, TPST2 loss-of-function emerged as a potential enhancer of anti-PD1 treatment efficacy. TPST2 suppressed IFNγ signaling by sulfating IFNγ receptor 1 at Y397 residue, while its downregulation boosted IFNγ-mediated signaling and antigen presentation. Depletion of TPST2 in cancer cells augmented anti-PD1 antibody efficacy in syngeneic mouse tumor models by enhancing tumor-infiltrating lymphocytes. RNA sequencing data revealed TPST2’s inverse correlation with antigen presentation, and increased TPST2 expression is associated with poor prognosis and altered cancer immunity across cancer types. We propose TPST2’s novel role as a suppressor of cancer immunity and advocate for its consideration as a therapeutic target in ICT-based treatments.
免疫检查点疗法(ICT)为部分癌症患者提供了持久的治疗效果,但耐药性仍然是一个重大挑战,这促使人们探索其潜在的分子机制。酪氨酸蛋白磺基转移酶-2(TPST2)因其在蛋白酪氨酸O-硫酸化中的作用而闻名,有人认为它能调节细胞外蛋白与蛋白之间的相互作用,但它在癌症免疫中的具体作用在很大程度上仍未被探索。为了探索影响抗 PD1 反应性的肿瘤细胞内在因素,我们在接种了人类免疫细胞的人源化小鼠中进行了功能缺失基因筛选。通过评估 IFNγ 介导的靶基因诱导、STAT1 磷酸化、HLA 表达和细胞生长抑制,评估了癌细胞对干扰素-γ(IFNγ)的反应性。通过共免疫沉淀和质谱分析确定了 TPST2 的硫代酪氨酸修饰靶基因。利用小鼠合成肿瘤模型评估了 TPST2 抑制作用的体内效应,并通过大量和单细胞 RNA 测序分析进行了证实。通过体内全基因组CRISPR筛选,TPST2功能缺失成为抗PD1疗效的潜在增强因子。TPST2通过硫酸化IFNγ受体1的Y397残基抑制IFNγ信号传导,而下调TPST2则促进IFNγ介导的信号传导和抗原递呈。通过增强肿瘤浸润淋巴细胞,消耗癌细胞中的TPST2可提高抗PD1抗体在合成小鼠肿瘤模型中的疗效。RNA 测序数据揭示了 TPST2 与抗原递呈的反相关性,TPST2 表达的增加与预后不良和不同癌症类型的癌症免疫改变有关。我们提出了 TPST2 作为癌症免疫抑制因子的新作用,并主张将其作为基于 ICT 的治疗靶点。
{"title":"Genome-wide CRISPR screening identifies tyrosylprotein sulfotransferase-2 as a target for augmenting anti-PD1 efficacy","authors":"Yumi Oh, Sujeong Kim, Yunjae Kim, Hyun Kim, Dongjun Jang, Seungjae Shin, Soo-Jin Lee, Jiwon Kim, Sang Eun Lee, Jaeik Oh, Yoojin Yang, Dohee Kim, Hae Rim Jung, Sangjin Kim, Jihui Kim, Kyungchan Min, Beomki Cho, Hoseok Seo, Dohyun Han, Hansoo Park, Sung-Yup Cho","doi":"10.1186/s12943-024-02068-x","DOIUrl":"https://doi.org/10.1186/s12943-024-02068-x","url":null,"abstract":"Immune checkpoint therapy (ICT) provides durable responses in select cancer patients, yet resistance remains a significant challenge, prompting the exploration of underlying molecular mechanisms. Tyrosylprotein sulfotransferase-2 (TPST2), known for its role in protein tyrosine O-sulfation, has been suggested to modulate the extracellular protein-protein interactions, but its specific role in cancer immunity remains largely unexplored. To explore tumor cell-intrinsic factors influencing anti-PD1 responsiveness, we conducted a pooled loss-of-function genetic screen in humanized mice engrafted with human immune cells. The responsiveness of cancer cells to interferon-γ (IFNγ) was estimated by evaluating IFNγ-mediated induction of target genes, STAT1 phosphorylation, HLA expression, and cell growth suppression. The sulfotyrosine-modified target gene of TPST2 was identified by co-immunoprecipitation and mass spectrometry. The in vivo effects of TPST2 inhibition were evaluated using mouse syngeneic tumor models and corroborated by bulk and single-cell RNA sequencing analyses. Through in vivo genome-wide CRISPR screening, TPST2 loss-of-function emerged as a potential enhancer of anti-PD1 treatment efficacy. TPST2 suppressed IFNγ signaling by sulfating IFNγ receptor 1 at Y397 residue, while its downregulation boosted IFNγ-mediated signaling and antigen presentation. Depletion of TPST2 in cancer cells augmented anti-PD1 antibody efficacy in syngeneic mouse tumor models by enhancing tumor-infiltrating lymphocytes. RNA sequencing data revealed TPST2’s inverse correlation with antigen presentation, and increased TPST2 expression is associated with poor prognosis and altered cancer immunity across cancer types. We propose TPST2’s novel role as a suppressor of cancer immunity and advocate for its consideration as a therapeutic target in ICT-based treatments.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141877664","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cancer, metastasis, and the epigenome 癌症、转移和表观基因组
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-02 DOI: 10.1186/s12943-024-02069-w
Saurav Kiri, Tyrone Ryba
Cancer is the second leading cause of death worldwide and disease burden is expected to increase globally throughout the next several decades, with the majority of cancer-related deaths occurring in metastatic disease. Cancers exhibit known hallmarks that endow them with increased survival and proliferative capacities, frequently as a result of de-stabilizing mutations. However, the genomic features that resolve metastatic clones from primary tumors are not yet well-characterized, as no mutational landscape has been identified as predictive of metastasis. Further, many cancers exhibit no known mutation signature. This suggests a larger role for non-mutational genome re-organization in promoting cancer evolution and dissemination. In this review, we highlight current critical needs for understanding cell state transitions and clonal selection advantages for metastatic cancer cells. We examine links between epigenetic states, genome structure, and misregulation of tumor suppressors and oncogenes, and discuss how recent technologies for understanding domain-scale regulation have been leveraged for a more complete picture of oncogenic and metastatic potential.
癌症是全球第二大死因,预计在未来几十年内,全球的疾病负担将不断加重,与癌症相关的死亡大多发生在转移性疾病上。癌症表现出的已知特征使其具有更强的生存和增殖能力,这往往是去稳定突变的结果。然而,从原发肿瘤中分化出转移性克隆的基因组特征尚未得到很好的描述,因为还没有发现任何突变景观可以预测转移。此外,许多癌症没有已知的突变特征。这表明,非突变基因组重组在促进癌症进化和扩散方面发挥着更大的作用。在这篇综述中,我们强调了目前了解细胞状态转换和转移癌细胞克隆选择优势的关键需求。我们研究了表观遗传状态、基因组结构以及肿瘤抑制因子和致癌基因的误调控之间的联系,并讨论了如何利用最新技术了解域尺度调控,从而更全面地了解致癌和转移潜力。
{"title":"Cancer, metastasis, and the epigenome","authors":"Saurav Kiri, Tyrone Ryba","doi":"10.1186/s12943-024-02069-w","DOIUrl":"https://doi.org/10.1186/s12943-024-02069-w","url":null,"abstract":"Cancer is the second leading cause of death worldwide and disease burden is expected to increase globally throughout the next several decades, with the majority of cancer-related deaths occurring in metastatic disease. Cancers exhibit known hallmarks that endow them with increased survival and proliferative capacities, frequently as a result of de-stabilizing mutations. However, the genomic features that resolve metastatic clones from primary tumors are not yet well-characterized, as no mutational landscape has been identified as predictive of metastasis. Further, many cancers exhibit no known mutation signature. This suggests a larger role for non-mutational genome re-organization in promoting cancer evolution and dissemination. In this review, we highlight current critical needs for understanding cell state transitions and clonal selection advantages for metastatic cancer cells. We examine links between epigenetic states, genome structure, and misregulation of tumor suppressors and oncogenes, and discuss how recent technologies for understanding domain-scale regulation have been leveraged for a more complete picture of oncogenic and metastatic potential.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141877667","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Single-cell tumor heterogeneity landscape of hepatocellular carcinoma: unraveling the pro-metastatic subtype and its interaction loop with fibroblasts 肝细胞癌的单细胞肿瘤异质性图谱:揭示促转移亚型及其与成纤维细胞的相互作用环路
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-02 DOI: 10.1186/s12943-024-02062-3
De-Zhen Guo, Xin Zhang, Sen-Quan Zhang, Shi-Yu Zhang, Xiang-Yu Zhang, Jia-Yan Yan, San-Yuan Dong, Kai Zhu, Xin-Rong Yang, Jia Fan, Jian Zhou, Ao Huang
Tumor heterogeneity presents a formidable challenge in understanding the mechanisms driving tumor progression and metastasis. The heterogeneity of hepatocellular carcinoma (HCC) in cellular level is not clear. Integration analysis of single-cell RNA sequencing data and spatial transcriptomics data was performed. Multiple methods were applied to investigate the subtype of HCC tumor cells. The functional characteristics, translation factors, clinical implications and microenvironment associations of different subtypes of tumor cells were analyzed. The interaction of subtype and fibroblasts were analyzed. We established a heterogeneity landscape of HCC malignant cells by integrated 52 single-cell RNA sequencing data and 5 spatial transcriptomics data. We identified three subtypes in tumor cells, including ARG1+ metabolism subtype (Metab-subtype), TOP2A+ proliferation phenotype (Prol-phenotype), and S100A6+ pro-metastatic subtype (EMT-subtype). Enrichment analysis found that the three subtypes harbored different features, that is metabolism, proliferating, and epithelial-mesenchymal transition. Trajectory analysis revealed that both Metab-subtype and EMT-subtype originated from the Prol-phenotype. Translation factor analysis found that EMT-subtype showed exclusive activation of SMAD3 and TGF-β signaling pathway. HCC dominated by EMT-subtype cells harbored an unfavorable prognosis and a deserted microenvironment. We uncovered a positive loop between tumor cells and fibroblasts mediated by SPP1-CD44 and CCN2/TGF-β-TGFBR1 interaction pairs. Inhibiting CCN2 disrupted the loop, mitigated the transformation to EMT-subtype, and suppressed metastasis. By establishing a heterogeneity landscape of malignant cells, we identified a three-subtype classification in HCC. Among them, S100A6+ tumor cells play a crucial role in metastasis. Targeting the feedback loop between tumor cells and fibroblasts is a promising anti-metastatic strategy.
肿瘤的异质性给了解肿瘤进展和转移的驱动机制带来了巨大挑战。肝细胞癌(HCC)在细胞水平上的异质性尚不清楚。研究人员对单细胞 RNA 测序数据和空间转录组学数据进行了整合分析。应用多种方法研究了 HCC 肿瘤细胞的亚型。分析了不同亚型肿瘤细胞的功能特征、翻译因子、临床意义和微环境关联。分析了亚型与成纤维细胞的相互作用。我们通过整合 52 个单细胞 RNA 测序数据和 5 个空间转录组学数据,建立了 HCC 恶性细胞的异质性图谱。我们在肿瘤细胞中发现了三种亚型,包括ARG1+代谢亚型(Metab-subtype)、TOP2A+增殖表型(Prol-phenotype)和S100A6+促转移亚型(EMT-subtype)。富集分析发现,这三种亚型具有不同的特征,即新陈代谢、增殖和上皮-间质转化。轨迹分析表明,代谢亚型和 EMT 亚型都源自 Prol 表型。翻译因子分析发现,EMT亚型显示出SMAD3和TGF-β信号通路的独家激活。以 EMT 亚型细胞为主的 HCC 预后不良,微环境荒芜。我们发现了肿瘤细胞与成纤维细胞之间由 SPP1-CD44 和 CCN2/TGF-β-TGFBR1 相互作用对介导的正循环。抑制CCN2可破坏这一循环,减轻向EMT亚型的转化,并抑制转移。通过建立恶性细胞的异质性图谱,我们确定了HCC的三亚型分类。其中,S100A6+肿瘤细胞在转移中起着至关重要的作用。针对肿瘤细胞和成纤维细胞之间的反馈回路是一种很有前景的抗转移策略。
{"title":"Single-cell tumor heterogeneity landscape of hepatocellular carcinoma: unraveling the pro-metastatic subtype and its interaction loop with fibroblasts","authors":"De-Zhen Guo, Xin Zhang, Sen-Quan Zhang, Shi-Yu Zhang, Xiang-Yu Zhang, Jia-Yan Yan, San-Yuan Dong, Kai Zhu, Xin-Rong Yang, Jia Fan, Jian Zhou, Ao Huang","doi":"10.1186/s12943-024-02062-3","DOIUrl":"https://doi.org/10.1186/s12943-024-02062-3","url":null,"abstract":"Tumor heterogeneity presents a formidable challenge in understanding the mechanisms driving tumor progression and metastasis. The heterogeneity of hepatocellular carcinoma (HCC) in cellular level is not clear. Integration analysis of single-cell RNA sequencing data and spatial transcriptomics data was performed. Multiple methods were applied to investigate the subtype of HCC tumor cells. The functional characteristics, translation factors, clinical implications and microenvironment associations of different subtypes of tumor cells were analyzed. The interaction of subtype and fibroblasts were analyzed. We established a heterogeneity landscape of HCC malignant cells by integrated 52 single-cell RNA sequencing data and 5 spatial transcriptomics data. We identified three subtypes in tumor cells, including ARG1+ metabolism subtype (Metab-subtype), TOP2A+ proliferation phenotype (Prol-phenotype), and S100A6+ pro-metastatic subtype (EMT-subtype). Enrichment analysis found that the three subtypes harbored different features, that is metabolism, proliferating, and epithelial-mesenchymal transition. Trajectory analysis revealed that both Metab-subtype and EMT-subtype originated from the Prol-phenotype. Translation factor analysis found that EMT-subtype showed exclusive activation of SMAD3 and TGF-β signaling pathway. HCC dominated by EMT-subtype cells harbored an unfavorable prognosis and a deserted microenvironment. We uncovered a positive loop between tumor cells and fibroblasts mediated by SPP1-CD44 and CCN2/TGF-β-TGFBR1 interaction pairs. Inhibiting CCN2 disrupted the loop, mitigated the transformation to EMT-subtype, and suppressed metastasis. By establishing a heterogeneity landscape of malignant cells, we identified a three-subtype classification in HCC. Among them, S100A6+ tumor cells play a crucial role in metastasis. Targeting the feedback loop between tumor cells and fibroblasts is a promising anti-metastatic strategy.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141877605","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Translational modeling-based evidence for enhanced efficacy of standard-of-care drugs in combination with anti-microRNA-155 in non-small-cell lung cancer 基于转化模型的证据表明,在非小细胞肺癌治疗中,标准治疗药物与抗微生物RNA-155联用可增强疗效
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-02 DOI: 10.1186/s12943-024-02060-5
Prashant Dogra, Vrushaly Shinglot, Javier Ruiz-Ramírez, Joseph Cave, Joseph D. Butner, Carmine Schiavone, Dan G. Duda, Ahmed O. Kaseb, Caroline Chung, Eugene J. Koay, Vittorio Cristini, Bulent Ozpolat, George A. Calin, Zhihui Wang
Elevated microRNA-155 (miR-155) expression in non-small-cell lung cancer (NSCLC) promotes cisplatin resistance and negatively impacts treatment outcomes. However, miR-155 can also boost anti-tumor immunity by suppressing PD-L1 expression. Therapeutic targeting of miR-155 through its antagonist, anti-miR-155, has proven challenging due to its dual molecular effects. We developed a multiscale mechanistic model, calibrated with in vivo data and then extrapolated to humans, to investigate the therapeutic effects of nanoparticle-delivered anti-miR-155 in NSCLC, alone or in combination with standard-of-care drugs. Model simulations and analyses of the clinical scenario revealed that monotherapy with anti-miR-155 at a dose of 2.5 mg/kg administered once every three weeks has substantial anti-cancer activity. It led to a median progression-free survival (PFS) of 6.7 months, which compared favorably to cisplatin and immune checkpoint inhibitors. Further, we explored the combinations of anti-miR-155 with standard-of-care drugs, and found strongly synergistic two- and three-drug combinations. A three-drug combination of anti-miR-155, cisplatin, and pembrolizumab resulted in a median PFS of 13.1 months, while a two-drug combination of anti-miR-155 and cisplatin resulted in a median PFS of 11.3 months, which emerged as a more practical option due to its simple design and cost-effectiveness. Our analyses also provided valuable insights into unfavorable dose ratios for drug combinations, highlighting the need for optimizing dose regimens to prevent antagonistic effects. This work bridges the gap between preclinical development and clinical translation of anti-miR-155 and unravels the potential of anti-miR-155 combination therapies in NSCLC.
非小细胞肺癌(NSCLC)中微RNA-155(miR-155)表达的升高会促进顺铂耐药,并对治疗效果产生负面影响。然而,miR-155 还能通过抑制 PD-L1 的表达来增强抗肿瘤免疫力。由于 miR-155 的双重分子效应,通过其拮抗剂 anti-miR-155 来靶向治疗 miR-155 已被证明具有挑战性。我们开发了一个多尺度机理模型,通过体内数据进行校准,然后外推至人体,以研究纳米颗粒递送的抗-miR-155 在 NSCLC 中单独或与标准治疗药物联用的治疗效果。模型模拟和临床情景分析表明,每三周一次、剂量为2.5 mg/kg的抗miR-155单药疗法具有很强的抗癌活性。该疗法的中位无进展生存期(PFS)为6.7个月,与顺铂和免疫检查点抑制剂相比效果更佳。此外,我们还探索了抗miR-155与标准治疗药物的组合,并发现了具有强烈协同作用的两药和三药组合。抗-miR-155、顺铂和pembrolizumab的三药联合疗法的中位PFS为13.1个月,而抗-miR-155和顺铂的两药联合疗法的中位PFS为11.3个月。我们的分析还为药物组合的不利剂量比提供了有价值的见解,强调了优化剂量方案以防止拮抗作用的必要性。这项研究填补了抗miR-155临床前开发与临床转化之间的空白,揭示了抗miR-155联合疗法在NSCLC中的潜力。
{"title":"Translational modeling-based evidence for enhanced efficacy of standard-of-care drugs in combination with anti-microRNA-155 in non-small-cell lung cancer","authors":"Prashant Dogra, Vrushaly Shinglot, Javier Ruiz-Ramírez, Joseph Cave, Joseph D. Butner, Carmine Schiavone, Dan G. Duda, Ahmed O. Kaseb, Caroline Chung, Eugene J. Koay, Vittorio Cristini, Bulent Ozpolat, George A. Calin, Zhihui Wang","doi":"10.1186/s12943-024-02060-5","DOIUrl":"https://doi.org/10.1186/s12943-024-02060-5","url":null,"abstract":"Elevated microRNA-155 (miR-155) expression in non-small-cell lung cancer (NSCLC) promotes cisplatin resistance and negatively impacts treatment outcomes. However, miR-155 can also boost anti-tumor immunity by suppressing PD-L1 expression. Therapeutic targeting of miR-155 through its antagonist, anti-miR-155, has proven challenging due to its dual molecular effects. We developed a multiscale mechanistic model, calibrated with in vivo data and then extrapolated to humans, to investigate the therapeutic effects of nanoparticle-delivered anti-miR-155 in NSCLC, alone or in combination with standard-of-care drugs. Model simulations and analyses of the clinical scenario revealed that monotherapy with anti-miR-155 at a dose of 2.5 mg/kg administered once every three weeks has substantial anti-cancer activity. It led to a median progression-free survival (PFS) of 6.7 months, which compared favorably to cisplatin and immune checkpoint inhibitors. Further, we explored the combinations of anti-miR-155 with standard-of-care drugs, and found strongly synergistic two- and three-drug combinations. A three-drug combination of anti-miR-155, cisplatin, and pembrolizumab resulted in a median PFS of 13.1 months, while a two-drug combination of anti-miR-155 and cisplatin resulted in a median PFS of 11.3 months, which emerged as a more practical option due to its simple design and cost-effectiveness. Our analyses also provided valuable insights into unfavorable dose ratios for drug combinations, highlighting the need for optimizing dose regimens to prevent antagonistic effects. This work bridges the gap between preclinical development and clinical translation of anti-miR-155 and unravels the potential of anti-miR-155 combination therapies in NSCLC.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141877665","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Molecular Cancer
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
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