首页 > 最新文献

bioRxiv - Cancer Biology最新文献

英文 中文
Craters on the melanoma surface facilitate tumor-immune interactions and demonstrate pathologic response to checkpoint blockade in humans 黑色素瘤表面的凹坑有助于肿瘤与免疫的相互作用,并显示出人类对检查点阻断剂的病理反应
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.18.613595
Aya Ludin, Georgia L. Stirtz, Asaf Tal, Ajit J. Nirmal, Naomi Besson, Stephanie M. Jones, Kathleen L. Pfaff, Michael Manos, Sophia Liu, Irving Barrera, Qiyu Gong, Cecilia Pessoa Rodrigues, Aditi Sahu, Elizabeth Jerison, Joao V. Alessi, Biagio Ricciuti, Douglas S. Richardson, Jodi D. Weiss, Hadley M. Moreau, Meredith E. Stanhope, Alexander B. Afeyan, James Sefton, Wyatt D. McCall, Emily Formato, Song Yang, Yi Zhou, David P. Hoytema van Konijnenburg, Hannah L. Cole, Miguel Cordova, Liang Deng, Milind Rajadhyaksha, Stephen R. Quake, Mark M. Awad, Fei Chen, Peter K. Sorger, F. Stephen Hodi, Scott J. Rodig, George F. Murphy, Leonard I. Zon
Immunotherapy leads to cancer eradication despite the tumor's immunosuppressive environment. Here, we used extended long-term in-vivo imaging and high-resolution spatial transcriptomics of endogenous melanoma in zebrafish, and multiplex imaging of human melanoma, to identify domains that facilitate immune response during immunotherapy. We identified crater-shaped pockets at the margins of zebrafish and human melanoma, rich with beta-2 microglobulin (B2M) and antigen recognition molecules. The craters harbor the highest density of CD8+ T cells in the tumor. In zebrafish, CD8+ T cells formed prolonged interactions with melanoma cells within craters, characteristic of antigen recognition. Following immunostimulatory treatment, the craters enlarged and became the major site of activated CD8+ T cell accumulation and tumor killing that was B2M dependent. In humans, craters predicted immune response to ICB therapy, showing response better than high T cell infiltration. This marks craters as potential new diagnostic tool for immunotherapy success and targets to enhance ICB response.
尽管肿瘤处于免疫抑制环境中,免疫疗法仍能导致癌症的根除。在这里,我们利用对斑马鱼内源性黑色素瘤的长期体内成像和高分辨率空间转录组学,以及对人类黑色素瘤的多重成像,确定了在免疫治疗过程中促进免疫反应的区域。我们在斑马鱼和人类黑色素瘤的边缘发现了火山口状的凹陷,其中富含β-2微球蛋白(B2M)和抗原识别分子。陨石坑是肿瘤中 CD8+ T 细胞密度最高的地方。在斑马鱼体内,CD8+ T 细胞与嵴内的黑色素瘤细胞形成了长时间的相互作用,这是抗原识别的特征。经过免疫刺激治疗后,陨石坑扩大,成为活化的 CD8+ T 细胞聚集和肿瘤杀伤的主要部位,这种杀伤依赖于 B2M。在人体中,陨石坑可预测对 ICB 治疗的免疫反应,其反应优于高 T 细胞浸润。这标志着陨石坑可能成为免疫疗法成功与否的新诊断工具,并成为增强 ICB 反应的目标。
{"title":"Craters on the melanoma surface facilitate tumor-immune interactions and demonstrate pathologic response to checkpoint blockade in humans","authors":"Aya Ludin, Georgia L. Stirtz, Asaf Tal, Ajit J. Nirmal, Naomi Besson, Stephanie M. Jones, Kathleen L. Pfaff, Michael Manos, Sophia Liu, Irving Barrera, Qiyu Gong, Cecilia Pessoa Rodrigues, Aditi Sahu, Elizabeth Jerison, Joao V. Alessi, Biagio Ricciuti, Douglas S. Richardson, Jodi D. Weiss, Hadley M. Moreau, Meredith E. Stanhope, Alexander B. Afeyan, James Sefton, Wyatt D. McCall, Emily Formato, Song Yang, Yi Zhou, David P. Hoytema van Konijnenburg, Hannah L. Cole, Miguel Cordova, Liang Deng, Milind Rajadhyaksha, Stephen R. Quake, Mark M. Awad, Fei Chen, Peter K. Sorger, F. Stephen Hodi, Scott J. Rodig, George F. Murphy, Leonard I. Zon","doi":"10.1101/2024.09.18.613595","DOIUrl":"https://doi.org/10.1101/2024.09.18.613595","url":null,"abstract":"Immunotherapy leads to cancer eradication despite the tumor's immunosuppressive environment. Here, we used extended long-term in-vivo imaging and high-resolution spatial transcriptomics of endogenous melanoma in zebrafish, and multiplex imaging of human melanoma, to identify domains that facilitate immune response during immunotherapy. We identified crater-shaped pockets at the margins of zebrafish and human melanoma, rich with beta-2 microglobulin (B2M) and antigen recognition molecules. The craters harbor the highest density of CD8+ T cells in the tumor. In zebrafish, CD8+ T cells formed prolonged interactions with melanoma cells within craters, characteristic of antigen recognition. Following immunostimulatory treatment, the craters enlarged and became the major site of activated CD8+ T cell accumulation and tumor killing that was B2M dependent. In humans, craters predicted immune response to ICB therapy, showing response better than high T cell infiltration. This marks craters as potential new diagnostic tool for immunotherapy success and targets to enhance ICB response.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249844","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DNFE: Directed-network flow entropy for detecting the tipping points during biological processes DNFE:用于检测生物过程中临界点的定向网络流熵
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.18.613673
Xueqing Peng, Peiluan Li, Luonan Chen
There generally exists a critical state or tipping point from a stable state to another in dynamic biological processes, beyond which a significant qualitative transition occurs. Identifying this tipping point and its driving network is essential to prevent or delay catastrophic consequences. However, most traditional approaches based on undirected networks still suffer from the problem of the robustness and effectiveness when applied to high-dimensional small sample data, especially for single-cell data. To address this challenge, we developed a directed-network flow entropy (DNFE) method which can transform measured omics data into a directed network. This method is applicable to both single-cell RNA-sequencing (scRNA-seq) and bulk data. By applying this method to five real datasets, including three single-cell datasets and two bulk tumor datasets, the method can not only successfully detect the critical states as well as their dynamic network biomarkers, but also help explore regulatory relationships between genes. Numerical simulation indicates that the DNFE method is robust and superior to existing methods. Furthermore, DNFE has predicted active transcription factors (TFs), and further identified 'dark genes', which are usually overlooked by traditional methods.
在动态生物过程中,从一个稳定状态到另一个稳定状态通常存在一个临界状态或临界点,超过这个临界点就会发生重大的质变。确定这一临界点及其驱动网络对于防止或延缓灾难性后果至关重要。然而,大多数基于无向网络的传统方法在应用于高维小样本数据,尤其是单细胞数据时,仍然存在鲁棒性和有效性问题。为解决这一难题,我们开发了一种有向网络流熵(DNFE)方法,它能将测量的omics数据转化为有向网络。这种方法适用于单细胞 RNA 序列(scRNA-seq)和批量数据。通过将该方法应用于五个真实数据集,包括三个单细胞数据集和两个肿瘤大数据集,该方法不仅能成功检测临界状态及其动态网络生物标志物,还有助于探索基因之间的调控关系。数值模拟表明,DNFE 方法具有鲁棒性,优于现有方法。此外,DNFE 还预测了活跃的转录因子 (TF),并进一步发现了通常被传统方法忽略的 "暗基因"。
{"title":"DNFE: Directed-network flow entropy for detecting the tipping points during biological processes","authors":"Xueqing Peng, Peiluan Li, Luonan Chen","doi":"10.1101/2024.09.18.613673","DOIUrl":"https://doi.org/10.1101/2024.09.18.613673","url":null,"abstract":"There generally exists a critical state or tipping point from a stable state to another in dynamic biological processes, beyond which a significant qualitative transition occurs. Identifying this tipping point and its driving network is essential to prevent or delay catastrophic consequences. However, most traditional approaches based on undirected networks still suffer from the problem of the robustness and effectiveness when applied to high-dimensional small sample data, especially for single-cell data. To address this challenge, we developed a directed-network flow entropy (DNFE) method which can transform measured omics data into a directed network. This method is applicable to both single-cell RNA-sequencing (scRNA-seq) and bulk data. By applying this method to five real datasets, including three single-cell datasets and two bulk tumor datasets, the method can not only successfully detect the critical states as well as their dynamic network biomarkers, but also help explore regulatory relationships between genes. Numerical simulation indicates that the DNFE method is robust and superior to existing methods. Furthermore, DNFE has predicted active transcription factors (TFs), and further identified 'dark genes', which are usually overlooked by traditional methods.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250082","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Transcriptional program-based deciphering of the MET exon 14 skipping regulation network 基于转录程序的 MET 14 号外显子跳越调控网络解密
Pub Date : 2024-09-19 DOI: 10.1101/2024.09.13.612820
Marie-Jose Truong, Geoffrey Pawlak, Jean-Pascal Meneboo, Sheherazade Sebda, Marie Fernandes, Martin Figeac, Mohamed Elati, David Tulasne
The MET exon 14 skipping mutation (named METex14del) described in lung cancer leads to prolonged activation of signaling pathways and aberrant cell responses, but the link between HGF signaling and cell responses remains unclear. A putative regulatory network of influential regulators of target genes was constructed from the transcriptomes of lung cancer cell lines. Overlaying this reference network with transcriptomic data from METex14del-expressing cells, stimulated or not by HGF, revealed a major regulatory node consisting mainly of the transcription factors ETS1, FOSL1 and SMAD3. HGF activation of METex14del induced the phosphorylation of these master regulators and the expression of their predicted target genes in a RAS-ERK pathway-dependent manner. Furthermore, most of the transcription factors in the regulatory node are known regulators of epithelial-mesenchymal transition, consistent with their involvement in migration and invasion. New modeling with transcriptomic data from MEK inhibitor-treated METex14del cells validated the key role of RAS-ERK pathway regulators and their target genes in METex14del receptor activation. Thus, we report an original strategy to identify key transcriptional nodes associated with specific signaling pathways that may become novel therapeutic targets.
肺癌中的 MET 14 号外显子跳越突变(命名为 METex14del)会导致信号通路的长期激活和异常细胞反应,但 HGF 信号与细胞反应之间的联系仍不清楚。研究人员从肺癌细胞系的转录组中构建了一个对靶基因有影响的潜在调控网络。将该参考网络与表达 METex14del 的细胞(无论是否受到 HGF 刺激)的转录组数据进行叠加,发现了一个主要的调控节点,该节点主要由转录因子 ETS1、FOSL1 和 SMAD3 组成。HGF 对 METex14del 的激活诱导了这些主调控因子的磷酸化,并以依赖 RAS-ERK 通路的方式诱导了其预测靶基因的表达。此外,调节节点中的大多数转录因子都是已知的上皮-间充质转化调节因子,这与它们参与迁移和侵袭是一致的。利用 MEK 抑制剂处理过的 METex14del 细胞的转录组数据建立的新模型验证了 RAS-ERK 通路调节因子及其靶基因在 METex14del 受体激活中的关键作用。因此,我们报告了一种独创的策略,以确定与特定信号通路相关的关键转录节点,这些节点可能成为新的治疗靶点。
{"title":"Transcriptional program-based deciphering of the MET exon 14 skipping regulation network","authors":"Marie-Jose Truong, Geoffrey Pawlak, Jean-Pascal Meneboo, Sheherazade Sebda, Marie Fernandes, Martin Figeac, Mohamed Elati, David Tulasne","doi":"10.1101/2024.09.13.612820","DOIUrl":"https://doi.org/10.1101/2024.09.13.612820","url":null,"abstract":"The MET exon 14 skipping mutation (named METex14del) described in lung cancer leads to prolonged activation of signaling pathways and aberrant cell responses, but the link between HGF signaling and cell responses remains unclear. A putative regulatory network of influential regulators of target genes was constructed from the transcriptomes of lung cancer cell lines. Overlaying this reference network with transcriptomic data from METex14del-expressing cells, stimulated or not by HGF, revealed a major regulatory node consisting mainly of the transcription factors ETS1, FOSL1 and SMAD3. HGF activation of METex14del induced the phosphorylation of these master regulators and the expression of their predicted target genes in a RAS-ERK pathway-dependent manner. Furthermore, most of the transcription factors in the regulatory node are known regulators of epithelial-mesenchymal transition, consistent with their involvement in migration and invasion. New modeling with transcriptomic data from MEK inhibitor-treated METex14del cells validated the key role of RAS-ERK pathway regulators and their target genes in METex14del receptor activation. Thus, we report an original strategy to identify key transcriptional nodes associated with specific signaling pathways that may become novel therapeutic targets.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250083","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Monitoring of Cell-free Human Papillomavirus DNA in Metastatic or Recurrent Cervical Cancer: Clinical Significance and Treatment Implications 监测转移性或复发性宫颈癌患者的无细胞人乳头状瘤病毒 DNA:临床意义和治疗影响
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.16.613189
Zhuomin Yin, Tao Feng, Qing Xu, Wumin Dai, Maowei Ni, Juan Ni, Hanmei Lou
Purpose: Monitoring of circulating human papillomavirus (HPV) cell-free DNA (cfDNA) is a minimally invasive approach for surveillance in HPV-associated cancers, particularly cervical cancer. The aim of this study was to monitor circulating HPV cfDNA levels in patients with recurrent or metastatic cervical cancer during treatment and follow-up to assess the utility of HPV cfDNA as a tumor marker for disease surveillance and in guiding clinical treatment decisions. Experimental Design: In this prospective pilot observational study, levels of HPV cfDNA in serum samples from 28 patients with recurrent or metastatic HPV+ cervical cancer were measured via digital droplet polymerase chain reaction. Results for HPV cfDNA levels were matched to clinical outcomes and to serum levels of squamous cell carcinoma antigen (SCC-Ag) to assess the clinical potential of HPV cfDNA as a tumor marker. Results: HPV cfDNA was detected in all 28 patients (100%). Notably, median baseline HPV cfDNA levels varied according to the metastatic pattern in individual patients (P=0.019). Specifically, patients with a combined multiple-metastasis pattern had higher median baseline HPV cfDNA levels than patients with a single metastasis (P=0.003). All participants exhibited changes in HPV cfDNA levels over a median monitoring period of 2 months (range 0.3–16.9) before evaluations for treatment response or disease progression. Among 26 patients initially diagnosed with squamous cell cervical cancer, the positivity rate was 100% for HPV cfDNA and 69.2% for SCC-Ag (P=0.004, 95% confidence interval, 0–0.391). Among 20 patients longitudinally monitored for squamous cell cervical cancer, the concordance with changes in disease status was 90% for HPV cfDNA and 50% for SCC-Ag (P=0.014, 95% confidence interval, 0.022–0.621). Conclusions: HPV cfDNA is a promising tumor marker for HPV+ cervical cancer that offers advantages over SCC-Ag. In the context of precision medicine, HPV cfDNA is poised to play an increasingly pivotal role in monitoring treatment efficacy, providing valuable insights into disease progression, and guiding clinical decisions.
目的:监测循环人乳头瘤病毒(HPV)无细胞DNA(cfDNA)是监测HPV相关癌症(尤其是宫颈癌)的一种微创方法。本研究旨在监测复发性或转移性宫颈癌患者在治疗和随访期间的循环 HPV cfDNA 水平,以评估 HPV cfDNA 作为肿瘤标志物在疾病监测和指导临床治疗决策方面的效用。实验设计:在这项前瞻性试点观察研究中,通过数字液滴聚合酶链反应测量了28例复发性或转移性HPV+宫颈癌患者血清样本中的HPV cfDNA水平。HPV cfDNA 含量结果与临床结果和血清中鳞状细胞癌抗原 (SCC-Ag) 含量相匹配,以评估 HPV cfDNA 作为肿瘤标志物的临床潜力。研究结果所有28名患者(100%)都检测到了HPV cfDNA。值得注意的是,HPV cfDNA 的中位基线水平因患者的转移模式而异(P=0.019)。具体来说,合并多处转移模式的患者的 HPV cfDNA 中位基线水平高于单处转移的患者(P=0.003)。在评估治疗反应或疾病进展之前,所有参与者的 HPV cfDNA 水平在 2 个月的中位监测期内(范围 0.3-16.9)都出现了变化。在 26 名初步诊断为鳞状细胞宫颈癌的患者中,HPV cfDNA 阳性率为 100%,SCC-Ag 阳性率为 69.2%(P=0.004,95% 置信区间为 0-0.391)。在纵向监测的 20 名鳞状细胞宫颈癌患者中,HPV cfDNA 与疾病状态变化的一致性为 90%,SCC-Ag 为 50%(P=0.014,95% 置信区间为 0.022-0.621)。结论:HPV cfDNA与 SCC-Ag 相比,HPV cfDNA 是一种很有前景的 HPV+ 宫颈癌肿瘤标记物。在精准医疗的背景下,HPV cfDNA 将在监测治疗效果、深入了解疾病进展情况以及指导临床决策方面发挥越来越重要的作用。
{"title":"Monitoring of Cell-free Human Papillomavirus DNA in Metastatic or Recurrent Cervical Cancer: Clinical Significance and Treatment Implications","authors":"Zhuomin Yin, Tao Feng, Qing Xu, Wumin Dai, Maowei Ni, Juan Ni, Hanmei Lou","doi":"10.1101/2024.09.16.613189","DOIUrl":"https://doi.org/10.1101/2024.09.16.613189","url":null,"abstract":"Purpose: Monitoring of circulating human papillomavirus (HPV) cell-free DNA (cfDNA) is a minimally invasive approach for surveillance in HPV-associated cancers, particularly cervical cancer. The aim of this study was to monitor circulating HPV cfDNA levels in patients with recurrent or metastatic cervical cancer during treatment and follow-up to assess the utility of HPV cfDNA as a tumor marker for disease surveillance and in guiding clinical treatment decisions. Experimental Design: In this prospective pilot observational study, levels of HPV cfDNA in serum samples from 28 patients with recurrent or metastatic HPV<sup>+</sup> cervical cancer were measured via digital droplet polymerase chain reaction. Results for HPV cfDNA levels were matched to clinical outcomes and to serum levels of squamous cell carcinoma antigen (SCC-Ag) to assess the clinical potential of HPV cfDNA as a tumor marker. Results: HPV cfDNA was detected in all 28 patients (100%). Notably, median baseline HPV cfDNA levels varied according to the metastatic pattern in individual patients (<em>P</em>=0.019). Specifically, patients with a combined multiple-metastasis pattern had higher median baseline HPV cfDNA levels than patients with a single metastasis (<em>P</em>=0.003). All participants exhibited changes in HPV cfDNA levels over a median monitoring period of 2 months (range 0.3–16.9) before evaluations for treatment response or disease progression. Among 26 patients initially diagnosed with squamous cell cervical cancer, the positivity rate was 100% for HPV cfDNA and 69.2% for SCC-Ag (<em>P</em>=0.004, 95% confidence interval, 0–0.391). Among 20 patients longitudinally monitored for squamous cell cervical cancer, the concordance with changes in disease status was 90% for HPV cfDNA and 50% for SCC-Ag (<em>P</em>=0.014, 95% confidence interval, 0.022–0.621). Conclusions: HPV cfDNA is a promising tumor marker for HPV<sup>+</sup> cervical cancer that offers advantages over SCC-Ag. In the context of precision medicine, HPV cfDNA is poised to play an increasingly pivotal role in monitoring treatment efficacy, providing valuable insights into disease progression, and guiding clinical decisions.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249756","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ex-vivo mouse precision cut tumour slices for modelling hepatocellular carcinoma; A 3Rs solution for at-scale drug screening 用于肝细胞癌建模的活体小鼠精确切割肿瘤切片;大规模药物筛选的 3Rs 解决方案
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.16.613213
Amy L Collins, Keara Kirkness, Erik Ramon-Gil, Eleni Tzortzopoulou, Daniel Geh, Ranie Cameron, Saimir Luli, Eman Khurram, Daniel Storey, Hannah Paish, David McDonald, Andrew Filby, Lee A Borthwick, Fiona Oakley, Derek Mann, Jack Leslie
Disease modelling is vital for improving knowledge of disease mechanisms and for development of new therapeutic molecules and strategies. Modelling the intact living tumour microenvironment (TME) is increasingly considered to be vital not only for gaining a better understanding of the biology of cancer but for examining the efficacy of novel oncology drugs. To date, pre-clinical mouse models of cancer have represented the mainstay methodology for studying the evolving TME and for determining the effects of potential therapeutic molecules on tumour evolution and growth. Regarding drug screening, in vivo mouse models are expensive, require the use of large cohorts of mice and involve the administration of drugs with unknown toxicities to animals which often result in adverse effects that can cause animal suffering and the discontinuation of drug investigations. Hepatocellular carcinoma (HCC) is a primary cancer of the liver for which there is an urgent need for improved systemic treatments due to the disease usually being diagnosed at an advanced stage and current treatments having limited efficacy. To provide a practical solution to the screening of drugs for their likely efficacy in HCC we have developed an ex-vivo model in which orthotopic tumours are excised from the liver and subsequently processed to generate precision-cut tumour slices (PCTS) which provide an intact culture model of the HCC-TME. We describe simplified culture conditions that maintain the viability and metabolic activity of live PCTS which maintain the architecture, cellular complexity, drug sensitivity and responsiveness to immunotherapy of the original tumour. Importantly, we show that HCC derived PCTS can be miniaturised to 96-well scale and modified to express soluble luciferase, which in combination enabled non-destructive screening of a library of 26 drugs at two doses using just 5 tumours as the source for PCTS. This screen identified two small molecules, salinomycin and rottlerin, that have potent anti-tumour activities in HCC-PCTS and subsequently validated salinomycin as effective in vivo. In summary, we report a 3Rs (reduction, refinement and replacement) solution for study of HCC biology and for 96-well-scale screening of potential therapeutic agents in the context of an intact, metabolically active TME.
疾病建模对于增进对疾病机理的了解以及开发新的治疗分子和策略至关重要。人们越来越认为,对完整的活体肿瘤微环境(TME)进行建模不仅对更好地了解癌症生物学,而且对研究新型肿瘤药物的疗效都至关重要。迄今为止,临床前癌症小鼠模型一直是研究不断变化的肿瘤微环境以及确定潜在治疗分子对肿瘤演变和生长的影响的主要方法。在药物筛选方面,体内小鼠模型价格昂贵,需要使用大量小鼠,而且需要对动物施用毒性未知的药物,这往往会导致不良反应,给动物带来痛苦,并导致药物研究中止。肝细胞癌(HCC)是一种原发性肝癌,由于该病通常在晚期才被诊断出来,而目前的治疗方法疗效有限,因此迫切需要改进系统治疗方法。为了提供一种切实可行的解决方案来筛选对 HCC 有疗效的药物,我们开发了一种体外模型,即从肝脏中切除正位肿瘤,然后进行处理以生成精确切割的肿瘤切片 (PCTS),从而提供一个完整的 HCC-TME 培养模型。我们描述了简化的培养条件,这些条件能保持活体 PCTS 的存活率和代谢活性,并能保持原始肿瘤的结构、细胞复杂性、药物敏感性和对免疫疗法的反应性。重要的是,我们发现 HCC 衍生的 PCTS 可以微型化到 96 孔的规模,并能表达可溶性荧光素酶,这两种方法结合使用,只需使用 5 个肿瘤作为 PCTS 的来源,就能以两种剂量对 26 种药物进行无损筛选。这一筛选确定了两种小分子药物,即盐霉素和罗曲林,它们在 HCC-PCTS 中具有强效抗肿瘤活性,随后验证了盐霉素在体内的有效性。总之,我们报告了一种 3R(还原、提纯和替代)解决方案,用于研究 HCC 生物学,并在完整、代谢活跃的 TME 背景下对潜在治疗药物进行 96 井规模的筛选。
{"title":"Ex-vivo mouse precision cut tumour slices for modelling hepatocellular carcinoma; A 3Rs solution for at-scale drug screening","authors":"Amy L Collins, Keara Kirkness, Erik Ramon-Gil, Eleni Tzortzopoulou, Daniel Geh, Ranie Cameron, Saimir Luli, Eman Khurram, Daniel Storey, Hannah Paish, David McDonald, Andrew Filby, Lee A Borthwick, Fiona Oakley, Derek Mann, Jack Leslie","doi":"10.1101/2024.09.16.613213","DOIUrl":"https://doi.org/10.1101/2024.09.16.613213","url":null,"abstract":"Disease modelling is vital for improving knowledge of disease mechanisms and for development of new therapeutic molecules and strategies. Modelling the intact living tumour microenvironment (TME) is increasingly considered to be vital not only for gaining a better understanding of the biology of cancer but for examining the efficacy of novel oncology drugs. To date, pre-clinical mouse models of cancer have represented the mainstay methodology for studying the evolving TME and for determining the effects of potential therapeutic molecules on tumour evolution and growth. Regarding drug screening, in vivo mouse models are expensive, require the use of large cohorts of mice and involve the administration of drugs with unknown toxicities to animals which often result in adverse effects that can cause animal suffering and the discontinuation of drug investigations. Hepatocellular carcinoma (HCC) is a primary cancer of the liver for which there is an urgent need for improved systemic treatments due to the disease usually being diagnosed at an advanced stage and current treatments having limited efficacy. To provide a practical solution to the screening of drugs for their likely efficacy in HCC we have developed an ex-vivo model in which orthotopic tumours are excised from the liver and subsequently processed to generate precision-cut tumour slices (PCTS) which provide an intact culture model of the HCC-TME. We describe simplified culture conditions that maintain the viability and metabolic activity of live PCTS which maintain the architecture, cellular complexity, drug sensitivity and responsiveness to immunotherapy of the original tumour. Importantly, we show that HCC derived PCTS can be miniaturised to 96-well scale and modified to express soluble luciferase, which in combination enabled non-destructive screening of a library of 26 drugs at two doses using just 5 tumours as the source for PCTS. This screen identified two small molecules, salinomycin and rottlerin, that have potent anti-tumour activities in HCC-PCTS and subsequently validated salinomycin as effective in vivo. In summary, we report a 3Rs (reduction, refinement and replacement) solution for study of HCC biology and for 96-well-scale screening of potential therapeutic agents in the context of an intact, metabolically active TME.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250086","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MAPK/ERK signaling in gliomas modulates interferon responses, T cell recruitment, enhances tumor-microglia crosstalk, and drives immune checkpoint blockade efficacy. 胶质瘤中的 MAPK/ERK 信号调节干扰素反应和 T 细胞招募,增强肿瘤与小胶质细胞之间的串扰,并促进免疫检查点阻断剂的疗效。
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.11.612571
Kwang-Soo Kim, Junyi Zhang, Victor Andres Arrieta, Crismita Dmello, Elena Grabis, Karl Habashy, Joseph Duffy, Junfei Zhao, Andrew Gould, Li Chen, Jian Hu, Irina Balyasnikova, Dhan Chand, Dan Levey, Peter Canoll, Wenting Zhao, Peter Sims, Raul Rabadan, Surya Pandey, Bin Zhang, Catalina Lee-Chang, Dieter Henrik Heiland, Adam Mendel Sonabend
Background: Glioblastoma (GB) remains a formidable challenge in neuro-oncology, with immune checkpoint blockade (ICB) showing limited efficacy in unselected patients. We previously recently established that MAPK/ERK signaling is associated with overall survival following anti-PD-1 and anti-CTLA-4 treatment in recurrent GB. However, the causal relationship between MAPK/ERK signaling and susceptibility to ICB, as well as the mechanisms underlying this association, remain poorly understood. Method: We conducted in vivo kinome-wide CRISPR/Cas9 screenings in murine gliomas to identify key regulators of susceptibility to anti-PD-1 and CD8+ T cell responses and performed survival studies to validate the most relevant genes. Additionally, paired single-cell RNA-sequencing (scRNA-seq) with p-ERK staining, spatial transcriptomics on GB samples, and ex-vivo slice culture of a BRAFV600E mutant GB tumor treated with BRAFi/MEKi were used to determine the causal relationship between MAPK signaling, tumor cell immunogenicity, and modulation of microglia phenotype. Results: CRISPR/Cas9 screens identified the MAPK pathway, particularly the RAF-MEK-ERK pathway, as the most critical modulator of glioma susceptibility to CD8+ T cells, and anti-PD-1 across all kinases. Experimentally-induced ERK phosphorylation in gliomas enhanced survival with ICB treatment, led to durable anti-tumoral immunity upon re-challenge and memory T cell infiltration in long-term survivors. Elevated p-ERK in glioma cells correlated with increased interferon responses, antigen presentation and T cell infiltration in GB. Moreover, spatial transcriptomics and scRNA-seq analysis revealed the modulation of interferon responses by the MAPK/ERK pathway in BRAFV600E human GB cells with ERK1/2 knockout and in slice cultures of human BRAFV600E GB tissue. Notably, BRAFi/MEKi treatment disrupted the interaction between tumor cells and tumor-associated macrophages/microglia in slice cultures from BRAFV600E mutant GB. Conclusion: Our data indicate that the MAPK/ERK pathway is a critical regulator of GB cell susceptibility to anti-tumoral immunity, modulating interferon responses, and antigen-presentation in glioma cells, as well as tumor cell interaction with microglia. These findings not only elucidate the mechanistic underpinnings of immunotherapy resistance in GB but also highlight the MAPK/ERK pathway as a promising target for enhancing immunotherapeutic efficacy in this challenging malignancy.
背景:胶质母细胞瘤(GB)仍然是神经肿瘤学领域的一项艰巨挑战,免疫检查点阻断(ICB)在未经选择的患者中显示出有限的疗效。我们最近发现,MAPK/ERK 信号传导与复发性脑胶质细胞瘤患者接受抗 PD-1 和抗 CTLA-4 治疗后的总生存率有关。然而,MAPK/ERK 信号传导与易感性之间的因果关系以及这种关联的机制仍不甚明了。研究方法我们在小鼠胶质瘤中进行了体内激元全CRISPR/Cas9筛选,以确定抗PD-1和CD8+ T细胞反应易感性的关键调节因子,并进行了生存研究以验证最相关的基因。此外,该研究还利用配对的单细胞RNA测序(scRNA-seq)与p-ERK染色、GB样本的空间转录组学以及用BRAFi/MEKi治疗的BRAFV600E突变GB肿瘤的体外切片培养来确定MAPK信号传导、肿瘤细胞免疫原性和小胶质细胞表型调控之间的因果关系。结果CRISPR/Cas9筛选确定了MAPK通路,尤其是RAF-MEK-ERK通路,是胶质瘤对CD8+ T细胞和抗PD-1所有激酶易感性的最关键调节因子。通过实验诱导胶质瘤中的ERK磷酸化,可提高ICB治疗的存活率,在再次挑战时产生持久的抗肿瘤免疫力,并在长期存活者中产生记忆T细胞浸润。胶质瘤细胞中 p-ERK 的升高与 GB 中干扰素反应、抗原递呈和 T 细胞浸润的增加相关。此外,空间转录组学和 scRNA-seq 分析显示,在 ERK1/2 基因敲除的 BRAFV600E 人 GB 细胞和人 BRAFV600E GB 组织的切片培养物中,MAPK/ERK 通路对干扰素反应有调节作用。值得注意的是,在 BRAFV600E 突变 GB 切片培养物中,BRAFi/MEKi 治疗破坏了肿瘤细胞与肿瘤相关巨噬细胞/小胶质细胞之间的相互作用。结论我们的数据表明,MAPK/ERK 通路是 GB 细胞易受抗肿瘤免疫影响的关键调节因子,可调节干扰素反应、胶质瘤细胞的抗原递呈以及肿瘤细胞与小胶质细胞的相互作用。这些发现不仅阐明了脑胶质瘤免疫治疗耐药的机理基础,而且还强调了MAPK/ERK通路是提高这种具有挑战性的恶性肿瘤免疫治疗效果的一个有希望的靶点。
{"title":"MAPK/ERK signaling in gliomas modulates interferon responses, T cell recruitment, enhances tumor-microglia crosstalk, and drives immune checkpoint blockade efficacy.","authors":"Kwang-Soo Kim, Junyi Zhang, Victor Andres Arrieta, Crismita Dmello, Elena Grabis, Karl Habashy, Joseph Duffy, Junfei Zhao, Andrew Gould, Li Chen, Jian Hu, Irina Balyasnikova, Dhan Chand, Dan Levey, Peter Canoll, Wenting Zhao, Peter Sims, Raul Rabadan, Surya Pandey, Bin Zhang, Catalina Lee-Chang, Dieter Henrik Heiland, Adam Mendel Sonabend","doi":"10.1101/2024.09.11.612571","DOIUrl":"https://doi.org/10.1101/2024.09.11.612571","url":null,"abstract":"Background: Glioblastoma (GB) remains a formidable challenge in neuro-oncology, with immune checkpoint blockade (ICB) showing limited efficacy in unselected patients. We previously recently established that MAPK/ERK signaling is associated with overall survival following anti-PD-1 and anti-CTLA-4 treatment in recurrent GB. However, the causal relationship between MAPK/ERK signaling and susceptibility to ICB, as well as the mechanisms underlying this association, remain poorly understood. Method: We conducted in vivo kinome-wide CRISPR/Cas9 screenings in murine gliomas to identify key regulators of susceptibility to anti-PD-1 and CD8+ T cell responses and performed survival studies to validate the most relevant genes. Additionally, paired single-cell RNA-sequencing (scRNA-seq) with p-ERK staining, spatial transcriptomics on GB samples, and ex-vivo slice culture of a BRAFV600E mutant GB tumor treated with BRAFi/MEKi were used to determine the causal relationship between MAPK signaling, tumor cell immunogenicity, and modulation of microglia phenotype. Results: CRISPR/Cas9 screens identified the MAPK pathway, particularly the RAF-MEK-ERK pathway, as the most critical modulator of glioma susceptibility to CD8+ T cells, and anti-PD-1 across all kinases. Experimentally-induced ERK phosphorylation in gliomas enhanced survival with ICB treatment, led to durable anti-tumoral immunity upon re-challenge and memory T cell infiltration in long-term survivors. Elevated p-ERK in glioma cells correlated with increased interferon responses, antigen presentation and T cell infiltration in GB. Moreover, spatial transcriptomics and scRNA-seq analysis revealed the modulation of interferon responses by the MAPK/ERK pathway in BRAFV600E human GB cells with ERK1/2 knockout and in slice cultures of human BRAFV600E GB tissue. Notably, BRAFi/MEKi treatment disrupted the interaction between tumor cells and tumor-associated macrophages/microglia in slice cultures from BRAFV600E mutant GB. Conclusion: Our data indicate that the MAPK/ERK pathway is a critical regulator of GB cell susceptibility to anti-tumoral immunity, modulating interferon responses, and antigen-presentation in glioma cells, as well as tumor cell interaction with microglia. These findings not only elucidate the mechanistic underpinnings of immunotherapy resistance in GB but also highlight the MAPK/ERK pathway as a promising target for enhancing immunotherapeutic efficacy in this challenging malignancy.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142250085","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
YTHDF1 Facilitates Lung Adenocarcinoma Progression via Promotion of EEF1G Translation in a m6A-Dependent Manner YTHDF1 通过依赖 m6A 促进 EEF1G 翻译促进肺腺癌进展
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.13.612607
Lihong Wang, Qihong Sheng, Xiaoyu Wang, Hongjuan Yue, Qian Wang, Mei Zhang, Junling Ma, Ling Wu, Jiaojiao Zhang, Zishuo Cheng, Weifang Yu, Ting Liu, Jia Wang
Lung adenocarcinoma (LUAD) is a malignant tumor with high morbidity and mortality worldwide, and overall survival rates for LUAD patients remain unimproved. RNA modification is a key process in post-transcriptional gene regulation in epigenetics, with N6-methyladenosine (m6A) being a common RNA modification. The molecular mechanisms of LUAD are unclear, but evidence suggests that m6A RNA methylation plays a significant role. This study aimed to clarify the role of YTHDF1 in LUAD development and pathogenesis. These findings confirmed that YTHDF1, a m6A reader protein, is highly expressed in LUAD tissues and is correlated with tumor differentiation and TNM stage. The results of functional loss experiments in LUAD cell lines revealed that downregulating YTHDF1 inhibits proliferation, migration, and invasion and induces apoptosis, with opposite effects observed upon YTHDF1 upregulation. In vivo, YTHDF1 knockout suppressed LUAD xenograft growth. RNA-seq, MeRIP-seq, RIP-seq, and bioinformatics analyses identified EEF1G as a downstream target of YTHDF1 in LUAD, and high expression of EEF1G was confirmed. The interaction between YTHDF1 and EEF1G was validated through RIP-qPCR, Co-IP and Co-IF assays. The overexpression of EEF1G in LUAD cells partially counteracts the tumor suppression induced by YTHDF1 silencing, and the knockdown of EEF1G has the opposite effect, further confirming the regulatory relationship. In summary, this study describes a novel YTHDF1/EEF1G regulatory pathway in which YTHDF1 promotes LUAD progression by recognizing and binding to the m6A-modified mRNA of EEF1G, accelerating its translation, suggesting that YTHDF1 may be a potential biomarker and therapeutic target.
肺腺癌(LUAD)是全世界发病率和死亡率都很高的恶性肿瘤,LUAD 患者的总体生存率仍未得到改善。在表观遗传学中,RNA修饰是转录后基因调控的关键过程,N6-甲基腺苷(m6A)是一种常见的RNA修饰。LUAD的分子机制尚不清楚,但有证据表明,m6A RNA甲基化起着重要作用。本研究旨在阐明YTHDF1在LUAD的发展和发病机制中的作用。研究结果证实,m6A读取蛋白YTHDF1在LUAD组织中高表达,并与肿瘤分化和TNM分期相关。在LUAD细胞系中进行的功能缺失实验结果显示,下调YTHDF1可抑制增殖、迁移和侵袭,并诱导细胞凋亡,而上调YTHDF1则会产生相反的效果。在体内,YTHDF1敲除抑制了LUAD异种移植的生长。通过RNA-seq、MeRIP-seq、RIP-seq和生物信息学分析发现,EEF1G是YTHDF1在LUAD中的下游靶标,并证实了EEF1G的高表达。通过RIP-qPCR、Co-IP和Co-IF检测验证了YTHDF1和EEF1G之间的相互作用。EEF1G 在 LUAD 细胞中的过表达部分抵消了 YTHDF1 沉默诱导的肿瘤抑制作用,而 EEF1G 的敲除则产生了相反的效果,进一步证实了两者之间的调控关系。综上所述,本研究描述了一种新的YTHDF1/EEF1G调控途径,其中YTHDF1通过识别并结合EEF1G的m6A修饰mRNA,加速其翻译,从而促进LUAD的进展,提示YTHDF1可能是一种潜在的生物标记物和治疗靶点。
{"title":"YTHDF1 Facilitates Lung Adenocarcinoma Progression via Promotion of EEF1G Translation in a m6A-Dependent Manner","authors":"Lihong Wang, Qihong Sheng, Xiaoyu Wang, Hongjuan Yue, Qian Wang, Mei Zhang, Junling Ma, Ling Wu, Jiaojiao Zhang, Zishuo Cheng, Weifang Yu, Ting Liu, Jia Wang","doi":"10.1101/2024.09.13.612607","DOIUrl":"https://doi.org/10.1101/2024.09.13.612607","url":null,"abstract":"Lung adenocarcinoma (LUAD) is a malignant tumor with high morbidity and mortality worldwide, and overall survival rates for LUAD patients remain unimproved. RNA modification is a key process in post-transcriptional gene regulation in epigenetics, with N6-methyladenosine (m6A) being a common RNA modification. The molecular mechanisms of LUAD are unclear, but evidence suggests that m6A RNA methylation plays a significant role. This study aimed to clarify the role of YTHDF1 in LUAD development and pathogenesis. These findings confirmed that YTHDF1, a m6A reader protein, is highly expressed in LUAD tissues and is correlated with tumor differentiation and TNM stage. The results of functional loss experiments in LUAD cell lines revealed that downregulating YTHDF1 inhibits proliferation, migration, and invasion and induces apoptosis, with opposite effects observed upon YTHDF1 upregulation. In vivo, YTHDF1 knockout suppressed LUAD xenograft growth. RNA-seq, MeRIP-seq, RIP-seq, and bioinformatics analyses identified EEF1G as a downstream target of YTHDF1 in LUAD, and high expression of EEF1G was confirmed. The interaction between YTHDF1 and EEF1G was validated through RIP-qPCR, Co-IP and Co-IF assays. The overexpression of EEF1G in LUAD cells partially counteracts the tumor suppression induced by YTHDF1 silencing, and the knockdown of EEF1G has the opposite effect, further confirming the regulatory relationship. In summary, this study describes a novel YTHDF1/EEF1G regulatory pathway in which YTHDF1 promotes LUAD progression by recognizing and binding to the m6A-modified mRNA of EEF1G, accelerating its translation, suggesting that YTHDF1 may be a potential biomarker and therapeutic target.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249759","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrative multiomic approaches reveal ZMAT3 and p21 as conserved hubs in the p53 tumor suppression network 多组学整合方法揭示 ZMAT3 和 p21 是 p53 肿瘤抑制网络中的保守枢纽
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.17.612743
Anthony M. Boutelle, Aicha R. Mabene, David Yao, Haiqing Xu, Mengxiong Wang, Yuning J. Tang, Steven S. Lopez, Sauradeep Sinha, Janos Demeter, Ran Cheng, Brooks A. Benard, Liz J. Valente, Alexandros P. Drainas, Martin D. Fischer, Ravindra Majeti, Dmitri Petrov, Peter K Jackson, Fan Yang, Monte M Winslow, Michael C Bassik, Laura D. Attardi
TP53, the most frequently mutated gene in human cancer, encodes a transcriptional activator that induces myriad downstream target genes. Despite the importance of p53 in tumor suppression, the specific p53 target genes important for tumor suppression remain unclear. Recent studies have identified the p53-inducible gene Zmat3 as a critical effector of tumor suppression, but many questions remain regarding its p53-dependence, activity across contexts, and mechanism of tumor suppression alone and in cooperation with other p53-inducible genes. To address these questions, we used Tuba-seqUltra somatic genome editing and tumor barcoding in a mouse lung adenocarcinoma model, combinatorial in vivo CRISPR/Cas9 screens, meta-analyses of gene expression and Cancer Dependency Map data, and integrative RNA-sequencing and shotgun proteomic analyses. We established Zmat3 as a core component of p53-mediated tumor suppression and identified Cdkn1a as the most potent cooperating p53-induced gene in tumor suppression. We discovered that ZMAT3/CDKN1A serve as near-universal effectors of p53-mediated tumor suppression that regulate cell division, migration, and extracellular matrix organization. Accordingly, combined Zmat3-Cdkn1a inactivation dramatically enhanced cell proliferation and migration compared to controls, akin to p53 inactivation. Together, our findings place ZMAT3 and CDKN1A as hubs of a p53-induced gene program that opposes tumorigenesis across various cellular and genetic contexts.
TP53 是人类癌症中最常见的突变基因,它编码的转录激活因子可诱导无数下游靶基因。尽管 p53 在抑制肿瘤方面起着重要作用,但对抑制肿瘤起重要作用的特定 p53 靶基因仍不清楚。最近的研究发现,p53 诱导的基因 Zmat3 是抑制肿瘤的关键效应因子,但关于它对 p53 的依赖性、在不同环境中的活性以及单独或与其他 p53 诱导的基因共同抑制肿瘤的机制仍存在许多问题。为了解决这些问题,我们在小鼠肺腺癌模型中使用了Tuba-seqUltra体细胞基因组编辑和肿瘤条形码、CRISPR/Cas9体内组合筛选、基因表达和癌症依赖图谱数据的荟萃分析以及RNA测序和散射蛋白质组的综合分析。我们确定 Zmat3 是 p53 介导的肿瘤抑制的核心成分,并确定 Cdkn1a 是 p53 诱导的肿瘤抑制中最有效的合作基因。我们发现,ZMAT3/CDKN1A几乎是p53介导的肿瘤抑制的普遍效应因子,可调节细胞分裂、迁移和细胞外基质组织。因此,与对照组相比,ZMAT3-CDKN1A联合失活会显著增强细胞增殖和迁移,这与p53失活类似。总之,我们的研究结果表明,ZMAT3 和 CDKN1A 是 p53 诱导的基因程序的枢纽,在各种细胞和基因环境中反对肿瘤发生。
{"title":"Integrative multiomic approaches reveal ZMAT3 and p21 as conserved hubs in the p53 tumor suppression network","authors":"Anthony M. Boutelle, Aicha R. Mabene, David Yao, Haiqing Xu, Mengxiong Wang, Yuning J. Tang, Steven S. Lopez, Sauradeep Sinha, Janos Demeter, Ran Cheng, Brooks A. Benard, Liz J. Valente, Alexandros P. Drainas, Martin D. Fischer, Ravindra Majeti, Dmitri Petrov, Peter K Jackson, Fan Yang, Monte M Winslow, Michael C Bassik, Laura D. Attardi","doi":"10.1101/2024.09.17.612743","DOIUrl":"https://doi.org/10.1101/2024.09.17.612743","url":null,"abstract":"<em>TP53</em>, the most frequently mutated gene in human cancer, encodes a transcriptional activator that induces myriad downstream target genes. Despite the importance of p53 in tumor suppression, the specific p53 target genes important for tumor suppression remain unclear. Recent studies have identified the p53-inducible gene <em>Zmat3</em> as a critical effector of tumor suppression, but many questions remain regarding its p53-dependence, activity across contexts, and mechanism of tumor suppression alone and in cooperation with other p53-inducible genes. To address these questions, we used Tuba-seq<sup>Ultra</sup> somatic genome editing and tumor barcoding in a mouse lung adenocarcinoma model, combinatorial <em>in vivo</em> CRISPR/Cas9 screens, meta-analyses of gene expression and Cancer Dependency Map data, and integrative RNA-sequencing and shotgun proteomic analyses. We established <em>Zmat3</em> as a core component of p53-mediated tumor suppression and identified <em>Cdkn1a</em> as the most potent cooperating p53-induced gene in tumor suppression. We discovered that <em>ZMAT3/CDKN1A</em> serve as near-universal effectors of p53-mediated tumor suppression that regulate cell division, migration, and extracellular matrix organization. Accordingly, combined <em>Zmat3-Cdkn1a</em> inactivation dramatically enhanced cell proliferation and migration compared to controls, akin to p53 inactivation. Together, our findings place <em>ZMAT3</em> and <em>CDKN1A</em> as hubs of a p53-induced gene program that opposes tumorigenesis across various cellular and genetic contexts.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249754","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of potent biparatopic antibodies targeting FGFR2 fusion driven cholangiocarcinoma. 鉴定针对 FGFR2 融合驱动的胆管癌的强效双抗体。
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.16.613045
Saireudee Chaturantabut, Sydney Oliver, Dennie T. Frederick, Jiwan Kim, Foxy P Robinson, Alessandro Sinopoli, Tian-Yu Song, Diego J Rodriguez, Liang Chang, Devishi Kesar, Yao He, Meilani Ching, Ruvimbo Dzvurumi, Adel Atari, Yuen-Yi Tseng, Nabeel Bardeesy, William R Sellers
Translocations involving FGFR2 gene fusions are common in cholangiocarcinoma and predict response to FGFR kinase inhibitors. However, the rate and durability of response are limited due to the emergence of resistance, typically involving acquired FGFR2 kinase domain mutations, and to sub-optimal dosing, relating to drug adverse effects. Here, we report the development of biparatopic antibodies targeting the FGFR2 extracellular domain (ECD), as candidate therapeutics. Biparatopic antibodies can overcome drawbacks of standard bivalent monoparatopic antibodies, which often show poor inhibitory or even agonist activity against oncogenic receptors. We show that oncogenic transformation by FGFR2 fusions requires an intact ECD. Moreover, by systematically generating biparatopic antibodies that target distinct epitope pairs along the FGFR2 ECD, we identified antibodies that effectively block signaling and malignant growth driven by FGFR2-fusions. Importantly, these antibodies demonstrate efficacy in vivo, synergy with FGFR inhibitors, and activity against FGFR2 fusions harboring kinase domain mutations. Thus, biparatopic antibodies may serve as new treatment options for patients with FGFR2-altered cholangiocarcinoma.
涉及 FGFR2 基因融合的易位在胆管癌中很常见,可预测对 FGFR 激酶抑制剂的反应。然而,由于耐药性的出现(通常涉及获得性 FGFR2 激酶域突变)以及与药物不良反应有关的次优剂量,反应的速率和持久性受到了限制。在此,我们报告了针对表皮生长因子受体2胞外结构域(ECD)的双位点抗体作为候选疗法的开发情况。双配位抗体可以克服标准双价单配位抗体的缺点,标准双价单配位抗体通常对致癌受体的抑制甚至激动活性较差。我们的研究表明,FGFR2 融合体的致癌转化需要完整的 ECD。此外,通过系统地生成以 FGFR2 ECD 上不同表位对为靶点的双靶抗体,我们发现了能有效阻断 FGFR2 融合体信号传导和恶性生长的抗体。重要的是,这些抗体在体内具有疗效,能与表皮生长因子受体抑制剂产生协同作用,并对激酶域突变的表皮生长因子受体2融合具有活性。因此,双抗体可作为FGFR2改变胆管癌患者的新治疗方案。
{"title":"Identification of potent biparatopic antibodies targeting FGFR2 fusion driven cholangiocarcinoma.","authors":"Saireudee Chaturantabut, Sydney Oliver, Dennie T. Frederick, Jiwan Kim, Foxy P Robinson, Alessandro Sinopoli, Tian-Yu Song, Diego J Rodriguez, Liang Chang, Devishi Kesar, Yao He, Meilani Ching, Ruvimbo Dzvurumi, Adel Atari, Yuen-Yi Tseng, Nabeel Bardeesy, William R Sellers","doi":"10.1101/2024.09.16.613045","DOIUrl":"https://doi.org/10.1101/2024.09.16.613045","url":null,"abstract":"Translocations involving FGFR2 gene fusions are common in cholangiocarcinoma and predict response to FGFR kinase inhibitors. However, the rate and durability of response are limited due to the emergence of resistance, typically involving acquired FGFR2 kinase domain mutations, and to sub-optimal dosing, relating to drug adverse effects. Here, we report the development of biparatopic antibodies targeting the FGFR2 extracellular domain (ECD), as candidate therapeutics. Biparatopic antibodies can overcome drawbacks of standard bivalent monoparatopic antibodies, which often show poor inhibitory or even agonist activity against oncogenic receptors. We show that oncogenic transformation by FGFR2 fusions requires an intact ECD. Moreover, by systematically generating biparatopic antibodies that target distinct epitope pairs along the FGFR2 ECD, we identified antibodies that effectively block signaling and malignant growth driven by FGFR2-fusions. Importantly, these antibodies demonstrate efficacy in vivo, synergy with FGFR inhibitors, and activity against FGFR2 fusions harboring kinase domain mutations. Thus, biparatopic antibodies may serve as new treatment options for patients with FGFR2-altered cholangiocarcinoma.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249755","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mutant p53 Misfolding and Aggregation Precedes Transformation into High-Grade Serous Ovarian Carcinoma 突变 p53 发生错折叠和聚集是转化为高级别浆液性卵巢癌的先兆
Pub Date : 2024-09-18 DOI: 10.1101/2024.09.17.612958
Sara Sartini, Lexi Omholt, Neda Moatamed, Alice Soragni
High Grade Serous Ovarian Cancer (HG-SOC), the most prevalent and aggressive gyneco-logical malignancy, is marked by ubiquitous loss of functional p53, largely due to point mutations that arise very early in carcinogenesis. These mu-tations often lead to p53 protein misfolding and subsequent aggregation, yet the alterations in in-tracellular p53 dynamics throughout ovarian can-cer progression remain poorly understood. HG-SOC originates from the fallopian tube epithelium, with a well-documented stepwise progression be-ginning with early pre-malignant p53 signatures. These signatures represent largely normal cells that express and accumulate mutant p53, which then transform into benign serous tubal intraepi-thelial lesions (STIL), progress into late pre-malig-nant serous tubal intraepithelial carcinoma (STIC), and ultimately lead to HGSOC. Here, we show that the transition from folded, soluble to aggregated mutant p53 occurs during the malignant transfor-mation of benign precursor lesions into HGSOC. We analyzed fallopian tube tissue collected from ten salpingo-oophorectomy cases and determined the proportion of cells carrying soluble versus mis-folded/mutant p53 through conformation-sensitive staining and quantification. Misfolded p53 protein, prone to aggregation, is present in STICs and HG-SOCs, but notably absent from pre-neoplastic le-sions and surrounding healthy tissue. Overall, our results indicate that aggregation of mutant p53 is a structural defect that distinguishes pre-neoplastic early lesions from late pre-malignant and malig-nant ones, offering a potential treatment window for targeting p53 aggregation and halting ovarian cancer progression.
高级别浆液性卵巢癌(HG-SOC)是发病率最高、侵袭性最强的妇科恶性肿瘤,其特征是功能性 p53 的普遍缺失,这主要是由于在癌变早期出现的点突变所致。这些突变通常会导致 p53 蛋白的错误折叠和随后的聚集,但人们对整个卵巢癌进展过程中细胞内 p53 动态的改变仍然知之甚少。HG-SOC 起源于输卵管上皮细胞,从早期恶性前的 p53 标志开始,逐步发展,这一点已得到充分证实。这些特征代表了表达和积累突变 p53 的大部分正常细胞,它们随后转变为良性浆液性输卵管上皮内病变(STIL),发展为晚期恶性前浆液性输卵管上皮内癌(STIC),并最终导致 HGSOC。在这里,我们发现,在良性前病变恶性转化为 HGSOC 的过程中,会发生突变 p53 从折叠、可溶到聚集的转变。我们分析了从十例输卵管切除术病例中收集的输卵管组织,并通过构象敏感染色和定量分析确定了携带可溶性与错误折叠/突变 p53 的细胞比例。易发生聚集的错误折叠 p53 蛋白存在于 STICs 和 HG-SOCs 中,但在肿瘤前浸润和周围健康组织中明显缺乏。总之,我们的研究结果表明,突变 p53 的聚集是一种结构缺陷,可将肿瘤前早期病变与晚期恶性前病变和恶性肿瘤区分开来,从而为靶向 p53 聚集和阻止卵巢癌进展提供了一个潜在的治疗窗口。
{"title":"Mutant p53 Misfolding and Aggregation Precedes Transformation into High-Grade Serous Ovarian Carcinoma","authors":"Sara Sartini, Lexi Omholt, Neda Moatamed, Alice Soragni","doi":"10.1101/2024.09.17.612958","DOIUrl":"https://doi.org/10.1101/2024.09.17.612958","url":null,"abstract":"High Grade Serous Ovarian Cancer (HG-SOC), the most prevalent and aggressive gyneco-logical malignancy, is marked by ubiquitous loss of functional p53, largely due to point mutations that arise very early in carcinogenesis. These mu-tations often lead to p53 protein misfolding and subsequent aggregation, yet the alterations in in-tracellular p53 dynamics throughout ovarian can-cer progression remain poorly understood. HG-SOC originates from the fallopian tube epithelium, with a well-documented stepwise progression be-ginning with early pre-malignant p53 signatures. These signatures represent largely normal cells that express and accumulate mutant p53, which then transform into benign serous tubal intraepi-thelial lesions (STIL), progress into late pre-malig-nant serous tubal intraepithelial carcinoma (STIC), and ultimately lead to HGSOC. Here, we show that the transition from folded, soluble to aggregated mutant p53 occurs during the malignant transfor-mation of benign precursor lesions into HGSOC. We analyzed fallopian tube tissue collected from ten salpingo-oophorectomy cases and determined the proportion of cells carrying soluble versus mis-folded/mutant p53 through conformation-sensitive staining and quantification. Misfolded p53 protein, prone to aggregation, is present in STICs and HG-SOCs, but notably absent from pre-neoplastic le-sions and surrounding healthy tissue. Overall, our results indicate that aggregation of mutant p53 is a structural defect that distinguishes pre-neoplastic early lesions from late pre-malignant and malig-nant ones, offering a potential treatment window for targeting p53 aggregation and halting ovarian cancer progression.","PeriodicalId":501233,"journal":{"name":"bioRxiv - Cancer Biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142249753","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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
bioRxiv - Cancer Biology
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