首页 > 最新文献

NPJ Precision Oncology最新文献

英文 中文
Cross-modal deep learning model for predicting pathologic complete response to neoadjuvant chemotherapy in breast cancer 预测乳腺癌新辅助化疗病理完全反应的跨模态深度学习模型。
IF 6.8 1区 医学 Q1 ONCOLOGY Pub Date : 2024-09-05 DOI: 10.1038/s41698-024-00678-8
Jianming Guo, Baihui Chen, Hongda Cao, Quan Dai, Ling Qin, Jinfeng Zhang, Youxue Zhang, Huanyu Zhang, Yuan Sui, Tianyu Chen, Dongxu Yang, Xue Gong, Dalin Li
Pathological complete response (pCR) serves as a critical measure of the success of neoadjuvant chemotherapy (NAC) in breast cancer, directly influencing subsequent therapeutic decisions. With the continuous advancement of artificial intelligence, methods for early and accurate prediction of pCR are being extensively explored. In this study, we propose a cross-modal multi-pathway automated prediction model that integrates temporal and spatial information. This model fuses digital pathology images from biopsy specimens and multi-temporal ultrasound (US) images to predict pCR status early in NAC. The model demonstrates exceptional predictive efficacy. Our findings lay the foundation for developing personalized treatment paradigms based on individual responses. This approach has the potential to become a critical auxiliary tool for the early prediction of NAC response in breast cancer patients.
病理完全反应(pCR)是衡量乳腺癌新辅助化疗(NAC)成功与否的关键指标,直接影响后续的治疗决策。随着人工智能的不断进步,人们正在广泛探索早期准确预测 pCR 的方法。在本研究中,我们提出了一种整合了时间和空间信息的跨模态多途径自动预测模型。该模型融合了活检标本的数字病理图像和多时相超声(US)图像,可预测 NAC 早期的 pCR 状态。该模型显示出卓越的预测功效。我们的研究结果为开发基于个体反应的个性化治疗范例奠定了基础。这种方法有望成为早期预测乳腺癌患者 NAC 反应的重要辅助工具。
{"title":"Cross-modal deep learning model for predicting pathologic complete response to neoadjuvant chemotherapy in breast cancer","authors":"Jianming Guo, Baihui Chen, Hongda Cao, Quan Dai, Ling Qin, Jinfeng Zhang, Youxue Zhang, Huanyu Zhang, Yuan Sui, Tianyu Chen, Dongxu Yang, Xue Gong, Dalin Li","doi":"10.1038/s41698-024-00678-8","DOIUrl":"10.1038/s41698-024-00678-8","url":null,"abstract":"Pathological complete response (pCR) serves as a critical measure of the success of neoadjuvant chemotherapy (NAC) in breast cancer, directly influencing subsequent therapeutic decisions. With the continuous advancement of artificial intelligence, methods for early and accurate prediction of pCR are being extensively explored. In this study, we propose a cross-modal multi-pathway automated prediction model that integrates temporal and spatial information. This model fuses digital pathology images from biopsy specimens and multi-temporal ultrasound (US) images to predict pCR status early in NAC. The model demonstrates exceptional predictive efficacy. Our findings lay the foundation for developing personalized treatment paradigms based on individual responses. This approach has the potential to become a critical auxiliary tool for the early prediction of NAC response in breast cancer patients.","PeriodicalId":19433,"journal":{"name":"NPJ Precision Oncology","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-09-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41698-024-00678-8.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142140685","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
M2 macrophage-derived lncRNA NORAD in EVs promotes NSCLC progression via miR-520g-3p/SMIM22/GALE axis EVs中源自M2巨噬细胞的lncRNA NORAD通过miR-520g-3p/SMIM22/GRE轴促进NSCLC的进展
IF 6.8 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-30 DOI: 10.1038/s41698-024-00675-x
Qingtao Zhao, Bin Li, Xiaopeng Zhang, Huanfen Zhao, Wenfei Xue, Zheng Yuan, Shun Xu, Guochen Duan
Non-small cell lung cancer (NSCLC) constitutes the majority of lung cancer cases, accounting for over 80%. RNAs in EVs play a pivotal role in various biological and pathological processes mediated by extracellular vesicle (EV). Long non-coding RNAs (lncRNAs) are widely associated with cancer-related functions, including cell proliferation, migration, invasion, and drug resistance. Tumor-associated macrophages are recognized as pivotal contributors to tumorigenesis. Given these insights, this study aims to uncover the impact of lncRNA NORAD in EVs derived from M2 macrophages in NSCLC cell lines and xenograft mouse models of NSCLC. EVs were meticulously isolated and verified based on their morphology and specific biomarkers. The interaction between lncRNA NORAD and SMIM22 was investigated using immunoprecipitation. The influence of SMIM22/GALE or lncRNA NORAD in EVs on glycolysis was assessed in NSCLC cell lines. Additionally, we evaluated the effects of M2 macrophage-derived lncRNA NORAD in EVs on cell proliferation and apoptosis through colony formation and flow cytometry assays. Furthermore, the impact of M2 macrophage-derived lncRNA NORAD in EVs on tumor growth was confirmed using xenograft tumor animal models. The results underscored the potential role of M2 macrophage-derived lncRNA NORAD in EVs in NSCLC. SMIM22/GALE promoted glycolysis and the proliferation of NSCLC cells. Furthermore, lncRNA NORAD in EVs targeted SMIM22 and miR-520g-3p in NSCLC cells. Notably, lncRNA NORAD in EVs promoted the proliferation of NSCLC cells and facilitated NSCLC tumor growth through the miR-520g-3p axis. In conclusion, M2 macrophage-derived lncRNA NORAD in EVs promotes NSCLC progression through the miR-520g-3p/SMIM22/GALE axis.
非小细胞肺癌(NSCLC)占肺癌病例的80%以上。EV中的RNA在细胞外囊泡介导的各种生物和病理过程中发挥着关键作用。长非编码 RNA(lncRNA)与癌症相关功能广泛相关,包括细胞增殖、迁移、侵袭和耐药性。肿瘤相关巨噬细胞被认为是肿瘤发生的关键因素。有鉴于此,本研究旨在揭示在NSCLC细胞系和NSCLC异种移植小鼠模型中从M2巨噬细胞提取的EVs中lncRNA NORAD的影响。研究人员根据EVs的形态和特定生物标志物对其进行了细致的分离和验证。利用免疫沉淀法研究了lncRNA NORAD与SMIM22之间的相互作用。在 NSCLC 细胞系中评估了 EVs 中的 SMIM22/GALE 或 lncRNA NORAD 对糖酵解的影响。此外,我们还通过集落形成和流式细胞术检测评估了EVs中M2巨噬细胞来源的lncRNA NORAD对细胞增殖和凋亡的影响。此外,我们还利用异种移植肿瘤动物模型证实了 EVs 中 M2 巨噬细胞衍生的 lncRNA NORAD 对肿瘤生长的影响。结果强调了EVs中M2巨噬细胞衍生的lncRNA NORAD在NSCLC中的潜在作用。SMIM22/GALE促进了NSCLC细胞的糖酵解和增殖。此外,EVs中的lncRNA NORAD靶向NSCLC细胞中的SMIM22和miR-520g-3p。值得注意的是,EVs中的lncRNA NORAD通过miR-520g-3p轴促进了NSCLC细胞的增殖,并促进了NSCLC肿瘤的生长。总之,EVs中来源于M2巨噬细胞的lncRNA NORAD通过miR-520g-3p/SMIM22/GALE轴促进NSCLC的进展。
{"title":"M2 macrophage-derived lncRNA NORAD in EVs promotes NSCLC progression via miR-520g-3p/SMIM22/GALE axis","authors":"Qingtao Zhao, Bin Li, Xiaopeng Zhang, Huanfen Zhao, Wenfei Xue, Zheng Yuan, Shun Xu, Guochen Duan","doi":"10.1038/s41698-024-00675-x","DOIUrl":"10.1038/s41698-024-00675-x","url":null,"abstract":"Non-small cell lung cancer (NSCLC) constitutes the majority of lung cancer cases, accounting for over 80%. RNAs in EVs play a pivotal role in various biological and pathological processes mediated by extracellular vesicle (EV). Long non-coding RNAs (lncRNAs) are widely associated with cancer-related functions, including cell proliferation, migration, invasion, and drug resistance. Tumor-associated macrophages are recognized as pivotal contributors to tumorigenesis. Given these insights, this study aims to uncover the impact of lncRNA NORAD in EVs derived from M2 macrophages in NSCLC cell lines and xenograft mouse models of NSCLC. EVs were meticulously isolated and verified based on their morphology and specific biomarkers. The interaction between lncRNA NORAD and SMIM22 was investigated using immunoprecipitation. The influence of SMIM22/GALE or lncRNA NORAD in EVs on glycolysis was assessed in NSCLC cell lines. Additionally, we evaluated the effects of M2 macrophage-derived lncRNA NORAD in EVs on cell proliferation and apoptosis through colony formation and flow cytometry assays. Furthermore, the impact of M2 macrophage-derived lncRNA NORAD in EVs on tumor growth was confirmed using xenograft tumor animal models. The results underscored the potential role of M2 macrophage-derived lncRNA NORAD in EVs in NSCLC. SMIM22/GALE promoted glycolysis and the proliferation of NSCLC cells. Furthermore, lncRNA NORAD in EVs targeted SMIM22 and miR-520g-3p in NSCLC cells. Notably, lncRNA NORAD in EVs promoted the proliferation of NSCLC cells and facilitated NSCLC tumor growth through the miR-520g-3p axis. In conclusion, M2 macrophage-derived lncRNA NORAD in EVs promotes NSCLC progression through the miR-520g-3p/SMIM22/GALE axis.","PeriodicalId":19433,"journal":{"name":"NPJ Precision Oncology","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41698-024-00675-x.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142091139","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
Pharmacogenomic discovery of genetically targeted cancer therapies optimized against clinical outcomes 根据临床结果优化基因靶向癌症疗法的药物基因组学发现
IF 6.8 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-28 DOI: 10.1038/s41698-024-00673-z
Peter Truesdell, Jessica Chang, Doris Coto Villa, Meiou Dai, Yulei Zhao, Robin McIlwain, Stephanie Young, Shawna Hiley, Andrew W. Craig, Tomas Babak
Despite the clinical success of dozens of genetically targeted cancer therapies, the vast majority of patients with tumors caused by loss-of-function (LoF) mutations do not have access to these treatments. This is primarily due to the challenge of developing a drug that treats a disease caused by the absence of a protein target. The success of PARP inhibitors has solidified synthetic lethality (SL) as a means to overcome this obstacle. Recent mapping of SL networks using pooled CRISPR-Cas9 screens is a promising approach for expanding this concept to treating cancers driven by additional LoF drivers. In practice, however, translating signals from cell lines, where these screens are typically conducted, to patient outcomes remains a challenge. We developed a pharmacogenomic (PGx) approach called “Clinically Optimized Driver Associated-PGx” (CODA-PGX) that accurately predicts genetically targeted therapies with clinical-stage efficacy in specific LoF driver contexts. Using approved targeted therapies and cancer drugs with available real-world evidence and molecular data from hundreds of patients, we discovered and optimized the key screening principles predictive of efficacy and overall patient survival. In addition to establishing basic technical conventions, such as drug concentration and screening kinetics, we found that replicating the driver perturbation in the right context, as well as selecting patients where those drivers are genuine founder mutations, were key to accurate translation. We used CODA-PGX to screen a diverse collection of clinical stage drugs and report dozens of novel LoF genetically targeted opportunities; many validated in xenografts and by real-world evidence. Notable examples include treating STAG2-mutant tumors with Carboplatin, SMARCB1-mutant tumors with Oxaliplatin, and TP53BP1-mutant tumors with Etoposide or Bleomycin. We identified principles of pharmacogenomic screening that predict clinical efficacy in cancer patients with specific driver mutations.
尽管数十种基因靶向癌症疗法取得了临床成功,但绝大多数因功能缺失(LoF)突变导致肿瘤的患者却无法获得这些治疗。这主要是由于开发一种药物来治疗因缺乏蛋白质靶点而导致的疾病所面临的挑战。PARP 抑制剂的成功巩固了合成致死(SL)作为克服这一障碍的手段的地位。最近利用集合 CRISPR-Cas9 筛选技术绘制的合成致死网络图是一种很有前景的方法,可将这一概念扩展到治疗由其他 LoF 驱动因素引起的癌症。然而,在实践中,如何将细胞系(这些筛选通常在细胞系中进行)的信号转化为患者的结果仍然是一个挑战。我们开发了一种名为 "临床优化驱动因素相关-PGX"(CODA-PGX)的药物基因组学(PGx)方法,它能准确预测在特定LoF驱动因素背景下具有临床阶段疗效的基因靶向疗法。我们利用已获批准的靶向疗法和抗癌药物以及来自数百名患者的现有实际证据和分子数据,发现并优化了预测疗效和患者总体生存期的关键筛选原则。除了建立基本的技术规范(如药物浓度和筛选动力学)外,我们还发现,在正确的背景下复制驱动因子扰动,以及选择驱动因子为真正创始突变的患者,是准确转化的关键。我们使用 CODA-PGX 筛选了一系列不同的临床阶段药物,并报告了数十个新的 LoF 基因靶向机会;其中许多已在异种移植中和现实世界中得到验证。显著的例子包括用卡铂治疗 STAG2 突变肿瘤,用奥沙利铂治疗 SMARCB1 突变肿瘤,用依托泊苷或博莱霉素治疗 TP53BP1 突变肿瘤。我们确定了药物基因组筛选的原则,这些原则可预测具有特定驱动基因突变的癌症患者的临床疗效。
{"title":"Pharmacogenomic discovery of genetically targeted cancer therapies optimized against clinical outcomes","authors":"Peter Truesdell, Jessica Chang, Doris Coto Villa, Meiou Dai, Yulei Zhao, Robin McIlwain, Stephanie Young, Shawna Hiley, Andrew W. Craig, Tomas Babak","doi":"10.1038/s41698-024-00673-z","DOIUrl":"10.1038/s41698-024-00673-z","url":null,"abstract":"Despite the clinical success of dozens of genetically targeted cancer therapies, the vast majority of patients with tumors caused by loss-of-function (LoF) mutations do not have access to these treatments. This is primarily due to the challenge of developing a drug that treats a disease caused by the absence of a protein target. The success of PARP inhibitors has solidified synthetic lethality (SL) as a means to overcome this obstacle. Recent mapping of SL networks using pooled CRISPR-Cas9 screens is a promising approach for expanding this concept to treating cancers driven by additional LoF drivers. In practice, however, translating signals from cell lines, where these screens are typically conducted, to patient outcomes remains a challenge. We developed a pharmacogenomic (PGx) approach called “Clinically Optimized Driver Associated-PGx” (CODA-PGX) that accurately predicts genetically targeted therapies with clinical-stage efficacy in specific LoF driver contexts. Using approved targeted therapies and cancer drugs with available real-world evidence and molecular data from hundreds of patients, we discovered and optimized the key screening principles predictive of efficacy and overall patient survival. In addition to establishing basic technical conventions, such as drug concentration and screening kinetics, we found that replicating the driver perturbation in the right context, as well as selecting patients where those drivers are genuine founder mutations, were key to accurate translation. We used CODA-PGX to screen a diverse collection of clinical stage drugs and report dozens of novel LoF genetically targeted opportunities; many validated in xenografts and by real-world evidence. Notable examples include treating STAG2-mutant tumors with Carboplatin, SMARCB1-mutant tumors with Oxaliplatin, and TP53BP1-mutant tumors with Etoposide or Bleomycin. We identified principles of pharmacogenomic screening that predict clinical efficacy in cancer patients with specific driver mutations.","PeriodicalId":19433,"journal":{"name":"NPJ Precision Oncology","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41698-024-00673-z.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142091189","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
Author Correction: TKI type switching overcomes ROS1 L2086F in ROS1 fusion-positive cancers 作者更正:TKI类型转换可克服ROS1融合阳性癌症中的ROS1 L2086F问题
IF 6.8 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-28 DOI: 10.1038/s41698-024-00676-w
Rajat Thawani, Matteo Repetto, Clare Keddy, Katelyn Nicholson, Kristen Jones, Kevin Nusser, Catherine Z. Beach, Guilherme Harada, Alexander Drilon, Monika A. Davare
{"title":"Author Correction: TKI type switching overcomes ROS1 L2086F in ROS1 fusion-positive cancers","authors":"Rajat Thawani, Matteo Repetto, Clare Keddy, Katelyn Nicholson, Kristen Jones, Kevin Nusser, Catherine Z. Beach, Guilherme Harada, Alexander Drilon, Monika A. Davare","doi":"10.1038/s41698-024-00676-w","DOIUrl":"10.1038/s41698-024-00676-w","url":null,"abstract":"","PeriodicalId":19433,"journal":{"name":"NPJ Precision Oncology","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-08-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41698-024-00676-w.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142091201","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
Targeting Nrf2/PHKG2 axis to enhance radiosensitivity in NSCLC 靶向 Nrf2/PHKG2 轴,提高 NSCLC 的放射敏感性。
IF 6.8 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-21 DOI: 10.1038/s41698-024-00629-3
Fushi Han, Shuzhen Chen, Kangwei Zhang, Kunming Zhang, Meng Wang, Peijun Wang
While ferroptosis shows promise in anti-cancer strategy, the molecular mechanisms behind this process remain poorly understood. Our research aims to highlight the regulation of radiotherapy-induced ferroptosis in non-small cell lung cancer (NSCLC) via the NRF2/PHKG2 axis-mediated mechanism. To identify ferroptosis-associated genes associated with radioresistance in NSCLC, this study employed high-throughput transcriptome sequencing and Lasso risk regression analysis. Clinical samples were analyzed to confirm PHKG2 expression changes before and after radiotherapy. The study further examined ferritinophagy-related factors, intracellular iron levels, mitochondrial function, and ferroptosis in NSCLC cells undergoing radiation exposure to explore the effect of PHKG2 on radiosensitivity or radioresistance. The research also demonstrated the transcriptional inhibition of PHKG2 by NRF2 and created in situ transplantation tumor models of NSCLC to examine the role of NRF2/PHKG2 axis in NSCLC radiosensitivity and resistance in vivo. The Lasso risk regression model that incorporated ferroptosis-associated genes effectively predicted the prognosis of patients with NSCLC. Radiotherapy-sensitive tissues exhibited an increased expression of PHKG2. Overexpression of PHKG2 led to elevated intracellular iron levels by promoting ferritinophagy and increased mitochondrial stress-dependent ferroptosis induced by radiotherapy. PHKG2 transcription repression was achieved through NRF2. The FAGs-Lasso risk regression model can accurately predict the prognosis of NSCLC patients. Targeting Nrf2 upregulates the expression of PHKG2 and reverses radiotherapy resistance in NSCLC by promoting iron autophagy and inducing mitochondrial dysfunction, thereby increasing radiotherapy sensitivity.
尽管铁突变有望成为抗癌策略,但人们对这一过程背后的分子机制仍然知之甚少。我们的研究旨在强调通过NRF2/PHKG2轴介导的机制调控放疗诱导的非小细胞肺癌(NSCLC)铁突变。为了鉴定与NSCLC放射耐药性相关的铁突变相关基因,本研究采用了高通量转录组测序和Lasso风险回归分析。对临床样本进行了分析,以确认放疗前后 PHKG2 表达的变化。研究进一步检测了接受放射照射的NSCLC细胞中嗜铁蛋白相关因子、细胞内铁水平、线粒体功能和铁突变,以探讨PHKG2对放射敏感性或放射抗性的影响。研究还证明了NRF2对PHKG2的转录抑制作用,并建立了NSCLC原位移植肿瘤模型,以研究NRF2/PHKG2轴在体内NSCLC放射敏感性和耐药性中的作用。纳入铁突变相关基因的Lasso风险回归模型能有效预测NSCLC患者的预后。放疗敏感组织的PHKG2表达量增加。PHKG2的过表达通过促进噬铁蛋白和增加放疗诱导的线粒体应激依赖性铁突变,导致细胞内铁水平升高。PHKG2 的转录抑制是通过 NRF2 实现的。FAGs-Lasso风险回归模型可准确预测NSCLC患者的预后。靶向Nrf2可上调PHKG2的表达,并通过促进铁自噬和诱导线粒体功能障碍逆转NSCLC的放疗耐药,从而提高放疗敏感性。
{"title":"Targeting Nrf2/PHKG2 axis to enhance radiosensitivity in NSCLC","authors":"Fushi Han, Shuzhen Chen, Kangwei Zhang, Kunming Zhang, Meng Wang, Peijun Wang","doi":"10.1038/s41698-024-00629-3","DOIUrl":"10.1038/s41698-024-00629-3","url":null,"abstract":"While ferroptosis shows promise in anti-cancer strategy, the molecular mechanisms behind this process remain poorly understood. Our research aims to highlight the regulation of radiotherapy-induced ferroptosis in non-small cell lung cancer (NSCLC) via the NRF2/PHKG2 axis-mediated mechanism. To identify ferroptosis-associated genes associated with radioresistance in NSCLC, this study employed high-throughput transcriptome sequencing and Lasso risk regression analysis. Clinical samples were analyzed to confirm PHKG2 expression changes before and after radiotherapy. The study further examined ferritinophagy-related factors, intracellular iron levels, mitochondrial function, and ferroptosis in NSCLC cells undergoing radiation exposure to explore the effect of PHKG2 on radiosensitivity or radioresistance. The research also demonstrated the transcriptional inhibition of PHKG2 by NRF2 and created in situ transplantation tumor models of NSCLC to examine the role of NRF2/PHKG2 axis in NSCLC radiosensitivity and resistance in vivo. The Lasso risk regression model that incorporated ferroptosis-associated genes effectively predicted the prognosis of patients with NSCLC. Radiotherapy-sensitive tissues exhibited an increased expression of PHKG2. Overexpression of PHKG2 led to elevated intracellular iron levels by promoting ferritinophagy and increased mitochondrial stress-dependent ferroptosis induced by radiotherapy. PHKG2 transcription repression was achieved through NRF2. The FAGs-Lasso risk regression model can accurately predict the prognosis of NSCLC patients. Targeting Nrf2 upregulates the expression of PHKG2 and reverses radiotherapy resistance in NSCLC by promoting iron autophagy and inducing mitochondrial dysfunction, thereby increasing radiotherapy sensitivity.","PeriodicalId":19433,"journal":{"name":"NPJ Precision Oncology","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41698-024-00629-3.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142018186","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
A synthetic cytotoxic T cell platform for rapidly prototyping TCR function 用于快速构建 TCR 功能原型的合成细胞毒性 T 细胞平台
IF 6.8 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-19 DOI: 10.1038/s41698-024-00669-9
Govinda Sharma, James Round, Fei Teng, Zahra Ali, Chris May, Eric Yung, Robert A. Holt
Current tools for functionally profiling T cell receptors with respect to cytotoxic potency and cross-reactivity are hampered by difficulties in establishing model systems to test these proteins in the contexts of different HLA alleles and against broad arrays of potential antigens. We have implemented a granzyme-activatable sensor of T cell cytotoxicity in a universal prototyping platform which enables facile recombinant expression of any combination of TCR-, peptide-, and class I MHC-coding sequences and direct assessment of resultant responses. This system consists of an engineered cell platform based on the immortalized natural killer cell line, YT-Indy, and the MHC-null antigen-presenting cell line, K562. These cells were engineered to furnish the YT-Indy/K562 pair with appropriate protein domains required for recombinant TCR expression and function in a non-T cell chassis, integrate a fluorescence-based target-centric early detection reporter of cytotoxic function, and deploy a set of protective genetic interventions designed to preserve antigen-presenting cells for subsequent capture and downstream characterization. Our data show successful reconstitution of the surface TCR complex in the YT-Indy cell line at biologically relevant levels. We also demonstrate successful induction and highly sensitive detection of antigen-specific response in multiple distinct model TCRs. Additionally, we monitored destruction of targets in co-culture and found that our survival-optimized system allowed for complete preservation after 24 h exposure to cytotoxic effectors. With this bioplatform, we anticipate investigators will be empowered to rapidly express and characterize T cell receptor responses, generate knowledge regarding the patterns of T cell receptor recognition, and optimize therapeutic T cell receptors.
由于难以建立模型系统来测试这些蛋白在不同 HLA 等位基因背景下以及针对广泛的潜在抗原阵列的作用,目前用于分析 T 细胞受体细胞毒性效力和交叉反应性的工具受到了阻碍。我们在一个通用的原型平台上实现了一种粒酶激活的 T 细胞细胞毒性传感器,它能方便地重组表达 TCR、多肽和 I 类 MHC 编码序列的任何组合,并直接评估由此产生的反应。该系统包括一个基于永生化自然杀伤细胞株 YT-Indy 和 MHC 缺失的抗原递呈细胞株 K562 的工程细胞平台。设计这些细胞的目的是为 YT-Indy/K562 细胞对提供在非 T 细胞底盘中重组 TCR 表达和功能所需的适当蛋白质结构域,整合基于荧光的以靶点为中心的细胞毒性功能早期检测报告器,并部署一套保护性遗传干预措施,旨在保留抗原递呈细胞,以便后续捕获和下游鉴定。我们的数据显示,YT-Indy 细胞系中的表面 TCR 复合物在生物相关水平上成功重组。我们还证明了在多个不同的模型 TCR 中成功诱导和高灵敏度地检测抗原特异性反应。此外,我们还监测了共培养中靶点的破坏情况,发现我们的生存优化系统可以在暴露于细胞毒性效应物 24 小时后完全保存靶点。有了这个生物平台,我们预计研究人员将有能力快速表达和描述 T 细胞受体反应,生成有关 T 细胞受体识别模式的知识,并优化治疗性 T 细胞受体。
{"title":"A synthetic cytotoxic T cell platform for rapidly prototyping TCR function","authors":"Govinda Sharma, James Round, Fei Teng, Zahra Ali, Chris May, Eric Yung, Robert A. Holt","doi":"10.1038/s41698-024-00669-9","DOIUrl":"10.1038/s41698-024-00669-9","url":null,"abstract":"Current tools for functionally profiling T cell receptors with respect to cytotoxic potency and cross-reactivity are hampered by difficulties in establishing model systems to test these proteins in the contexts of different HLA alleles and against broad arrays of potential antigens. We have implemented a granzyme-activatable sensor of T cell cytotoxicity in a universal prototyping platform which enables facile recombinant expression of any combination of TCR-, peptide-, and class I MHC-coding sequences and direct assessment of resultant responses. This system consists of an engineered cell platform based on the immortalized natural killer cell line, YT-Indy, and the MHC-null antigen-presenting cell line, K562. These cells were engineered to furnish the YT-Indy/K562 pair with appropriate protein domains required for recombinant TCR expression and function in a non-T cell chassis, integrate a fluorescence-based target-centric early detection reporter of cytotoxic function, and deploy a set of protective genetic interventions designed to preserve antigen-presenting cells for subsequent capture and downstream characterization. Our data show successful reconstitution of the surface TCR complex in the YT-Indy cell line at biologically relevant levels. We also demonstrate successful induction and highly sensitive detection of antigen-specific response in multiple distinct model TCRs. Additionally, we monitored destruction of targets in co-culture and found that our survival-optimized system allowed for complete preservation after 24 h exposure to cytotoxic effectors. With this bioplatform, we anticipate investigators will be empowered to rapidly express and characterize T cell receptor responses, generate knowledge regarding the patterns of T cell receptor recognition, and optimize therapeutic T cell receptors.","PeriodicalId":19433,"journal":{"name":"NPJ Precision Oncology","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41698-024-00669-9.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142002634","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
Biologically interpretable multi-task deep learning pipeline predicts molecular alterations, grade, and prognosis in glioma patients 从生物学角度解读多任务深度学习管道,预测胶质瘤患者的分子改变、分级和预后。
IF 6.8 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-16 DOI: 10.1038/s41698-024-00670-2
Xuewei Wu, Shuaitong Zhang, Zhenyu Zhang, Zicong He, Zexin Xu, Weiwei Wang, Zhe Jin, Jingjing You, Yang Guo, Lu Zhang, Wenhui Huang, Fei Wang, Xianzhi Liu, Dongming Yan, Jingliang Cheng, Jing Yan, Shuixing Zhang, Bin Zhang
Deep learning models have been developed for various predictions in glioma; yet, they were constrained by manual segmentation, task-specific design, or a lack of biological interpretation. Herein, we aimed to develop an end-to-end multi-task deep learning (MDL) pipeline that can simultaneously predict molecular alterations and histological grade (auxiliary tasks), as well as prognosis (primary task) in gliomas. Further, we aimed to provide the biological mechanisms underlying the model’s predictions. We collected multiscale data including baseline MRI images from 2776 glioma patients across two private (FAHZU and HPPH, n = 1931) and three public datasets (TCGA, n = 213; UCSF, n = 410; and EGD, n = 222). We trained and internally validated the MDL model using our private datasets, and externally validated it using the three public datasets. We used the model-predicted deep prognosis score (DPS) to stratify patients into low-DPS and high-DPS subtypes. Additionally, a radio-multiomics analysis was conducted to elucidate the biological basis of the DPS. In the external validation cohorts, the MDL model achieved average areas under the curve of 0.892–0.903, 0.710–0.894, and 0.850–0.879 for predicting IDH mutation status, 1p/19q co-deletion status, and tumor grade, respectively. Moreover, the MDL model yielded a C-index of 0.723 in the TCGA and 0.671 in the UCSF for the prediction of overall survival. The DPS exhibits significant correlations with activated oncogenic pathways, immune infiltration patterns, specific protein expression, DNA methylation, tumor mutation burden, and tumor-stroma ratio. Accordingly, our work presents an accurate and biologically meaningful tool for predicting molecular subtypes, tumor grade, and survival outcomes in gliomas, which provides personalized clinical decision-making in a global and non-invasive manner.
深度学习模型已被开发用于胶质瘤的各种预测;然而,它们受到手动分割、特定任务设计或缺乏生物学解释的限制。在此,我们旨在开发一种端到端多任务深度学习(MDL)管道,它可以同时预测胶质瘤的分子改变和组织学分级(辅助任务)以及预后(主要任务)。此外,我们还旨在提供该模型预测所依据的生物学机制。我们收集了多尺度数据,包括来自两个私人数据集(FAHZU 和 HPPH,n = 1931)和三个公共数据集(TCGA,n = 213;UCSF,n = 410;EGD,n = 222)的 2776 例胶质瘤患者的基线 MRI 图像。我们使用私有数据集对 MDL 模型进行了训练和内部验证,并使用三个公共数据集对其进行了外部验证。我们使用模型预测的深度预后评分(DPS)将患者分为低DPS亚型和高DPS亚型。此外,我们还进行了放射多组学分析,以阐明 DPS 的生物学基础。在外部验证队列中,MDL模型预测IDH突变状态、1p/19q共缺失状态和肿瘤分级的平均曲线下面积分别为0.892-0.903、0.710-0.894和0.850-0.879。此外,MDL 模型在 TCGA 和 UCSF 中预测总生存期的 C 指数分别为 0.723 和 0.671。DPS 与活化的致癌通路、免疫浸润模式、特异性蛋白表达、DNA 甲基化、肿瘤突变负荷和肿瘤-间质比率有明显的相关性。因此,我们的工作为预测胶质瘤的分子亚型、肿瘤分级和生存结果提供了一种准确且具有生物学意义的工具,它能以全局性和非侵入性的方式提供个性化的临床决策。
{"title":"Biologically interpretable multi-task deep learning pipeline predicts molecular alterations, grade, and prognosis in glioma patients","authors":"Xuewei Wu, Shuaitong Zhang, Zhenyu Zhang, Zicong He, Zexin Xu, Weiwei Wang, Zhe Jin, Jingjing You, Yang Guo, Lu Zhang, Wenhui Huang, Fei Wang, Xianzhi Liu, Dongming Yan, Jingliang Cheng, Jing Yan, Shuixing Zhang, Bin Zhang","doi":"10.1038/s41698-024-00670-2","DOIUrl":"10.1038/s41698-024-00670-2","url":null,"abstract":"Deep learning models have been developed for various predictions in glioma; yet, they were constrained by manual segmentation, task-specific design, or a lack of biological interpretation. Herein, we aimed to develop an end-to-end multi-task deep learning (MDL) pipeline that can simultaneously predict molecular alterations and histological grade (auxiliary tasks), as well as prognosis (primary task) in gliomas. Further, we aimed to provide the biological mechanisms underlying the model’s predictions. We collected multiscale data including baseline MRI images from 2776 glioma patients across two private (FAHZU and HPPH, n = 1931) and three public datasets (TCGA, n = 213; UCSF, n = 410; and EGD, n = 222). We trained and internally validated the MDL model using our private datasets, and externally validated it using the three public datasets. We used the model-predicted deep prognosis score (DPS) to stratify patients into low-DPS and high-DPS subtypes. Additionally, a radio-multiomics analysis was conducted to elucidate the biological basis of the DPS. In the external validation cohorts, the MDL model achieved average areas under the curve of 0.892–0.903, 0.710–0.894, and 0.850–0.879 for predicting IDH mutation status, 1p/19q co-deletion status, and tumor grade, respectively. Moreover, the MDL model yielded a C-index of 0.723 in the TCGA and 0.671 in the UCSF for the prediction of overall survival. The DPS exhibits significant correlations with activated oncogenic pathways, immune infiltration patterns, specific protein expression, DNA methylation, tumor mutation burden, and tumor-stroma ratio. Accordingly, our work presents an accurate and biologically meaningful tool for predicting molecular subtypes, tumor grade, and survival outcomes in gliomas, which provides personalized clinical decision-making in a global and non-invasive manner.","PeriodicalId":19433,"journal":{"name":"NPJ Precision Oncology","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11329669/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141996230","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
Probing the killing potency of tumor-infiltrating lymphocytes on microarrayed colorectal cancer tumoroids 探究肿瘤浸润淋巴细胞对微阵列结直肠癌肿瘤细胞的杀伤力
IF 6.8 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-14 DOI: 10.1038/s41698-024-00661-3
Devanjali Dutta, L. Francisco Lorenzo-Martín, François Rivest, Nicolas Broguiere, Lucie Tillard, Simone Ragusa, Nathalie Brandenberg, Sylke Höhnel, Damien Saugy, Sylvie Rusakiewicz, Krisztian Homicsko, George Coukos, Matthias P. Lutolf
Immunotherapy has emerged as a new standard of care for certain cancer patients with specific cellular and molecular makeups. However, there is still an unmet need for ex vivo models able to readily assess the effectiveness of immunotherapeutic treatments in a high-throughput and patient-specific manner. To address this issue, we have developed a microarrayed system of patient-derived tumoroids with recreated immune microenvironments that are optimized for the high-content evaluation of tumor-infiltrating lymphocyte functionality. Here we show that this system offers unprecedented opportunities to evaluate tumor immunogenicity, characterize the response to immunomodulators, and explore novel approaches for personalized immuno-oncology.
免疫疗法已成为治疗某些具有特定细胞和分子组成的癌症患者的新标准。然而,对于能够以高通量和患者特异性的方式随时评估免疫治疗效果的体外模型,我们仍有未满足的需求。为了解决这个问题,我们开发了一种患者来源肿瘤微阵列系统,该系统具有重新创建的免疫微环境,可优化肿瘤浸润淋巴细胞功能的高含量评估。我们在这里展示的这一系统为评估肿瘤免疫原性、描述对免疫调节剂的反应以及探索个性化免疫肿瘤学的新方法提供了前所未有的机会。
{"title":"Probing the killing potency of tumor-infiltrating lymphocytes on microarrayed colorectal cancer tumoroids","authors":"Devanjali Dutta, L. Francisco Lorenzo-Martín, François Rivest, Nicolas Broguiere, Lucie Tillard, Simone Ragusa, Nathalie Brandenberg, Sylke Höhnel, Damien Saugy, Sylvie Rusakiewicz, Krisztian Homicsko, George Coukos, Matthias P. Lutolf","doi":"10.1038/s41698-024-00661-3","DOIUrl":"10.1038/s41698-024-00661-3","url":null,"abstract":"Immunotherapy has emerged as a new standard of care for certain cancer patients with specific cellular and molecular makeups. However, there is still an unmet need for ex vivo models able to readily assess the effectiveness of immunotherapeutic treatments in a high-throughput and patient-specific manner. To address this issue, we have developed a microarrayed system of patient-derived tumoroids with recreated immune microenvironments that are optimized for the high-content evaluation of tumor-infiltrating lymphocyte functionality. Here we show that this system offers unprecedented opportunities to evaluate tumor immunogenicity, characterize the response to immunomodulators, and explore novel approaches for personalized immuno-oncology.","PeriodicalId":19433,"journal":{"name":"NPJ Precision Oncology","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11324658/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141982887","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
Comprehensive genetic profiling and molecularly guided treatment for patients with primary CNS tumors 对原发性中枢神经系统肿瘤患者进行全面基因分析和分子指导治疗。
IF 6.8 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-14 DOI: 10.1038/s41698-024-00674-y
Julia C. Kuehn, Patrick Metzger, Nicolas Neidert, Uta Matysiak, Linda Gräßel, Ulrike Philipp, Sabine Bleul, Thomas Pauli, Julia Falkenstein, Henriette Bertemes, Stepan Cysar, Maria Elena Hess, Anna Verena Frey, Jesús Duque-Afonso, Elisabeth Schorb, Marcia Machein, Jürgen Beck, Oliver Schnell, Nikolas von Bubnoff, Anna L. Illert, Christoph Peters, Tilman Brummer, Marco Prinz, Cornelius Miething, Heiko Becker, Silke Lassmann, Martin Werner, Melanie Börries, Justus Duyster, Dieter H. Heiland, Roman Sankowski, Florian Scherer
Despite major advances in molecular profiling and classification of primary brain tumors, personalized treatment remains limited for most patients. Here, we explored the feasibility of individual molecular profiling and the efficacy of biomarker-guided therapy for adult patients with primary brain cancers in the real-world setting within the molecular tumor board Freiburg, Germany. We analyzed genetic profiles, personalized treatment recommendations, and clinical outcomes of 102 patients with 21 brain tumor types. Alterations in the cell cycle, BRAF, and mTOR pathways most frequently led to personalized treatment recommendations. Molecularly informed therapies were recommended in 71% and implemented in 32% of patients with completed molecular diagnostics. The disease control rate following targeted treatment was 50% and the overall response rate was 30%, with a progression-free survival 2/1 ratio of at least 1.3 in 31% of patients. This study highlights the efficacy of molecularly guided treatment and the need for biomarker-stratified trials in brain cancers.
尽管在原发性脑肿瘤的分子图谱分析和分类方面取得了重大进展,但对大多数患者来说,个性化治疗仍然有限。在这里,我们探讨了在德国弗莱堡分子肿瘤委员会的真实世界环境中,对原发性脑癌成年患者进行个体分子图谱分析的可行性和生物标志物指导治疗的疗效。我们分析了 102 名 21 种脑肿瘤患者的基因图谱、个性化治疗建议和临床疗效。细胞周期、BRAF 和 mTOR 通路的改变最常导致个性化治疗建议。在完成分子诊断的患者中,71%的患者获得了分子信息疗法建议,32%的患者实施了分子信息疗法。靶向治疗后的疾病控制率为50%,总体反应率为30%,31%的患者无进展生存期2/1比率至少为1.3。这项研究凸显了分子引导治疗的疗效以及对脑癌生物标志物分层试验的需求。
{"title":"Comprehensive genetic profiling and molecularly guided treatment for patients with primary CNS tumors","authors":"Julia C. Kuehn, Patrick Metzger, Nicolas Neidert, Uta Matysiak, Linda Gräßel, Ulrike Philipp, Sabine Bleul, Thomas Pauli, Julia Falkenstein, Henriette Bertemes, Stepan Cysar, Maria Elena Hess, Anna Verena Frey, Jesús Duque-Afonso, Elisabeth Schorb, Marcia Machein, Jürgen Beck, Oliver Schnell, Nikolas von Bubnoff, Anna L. Illert, Christoph Peters, Tilman Brummer, Marco Prinz, Cornelius Miething, Heiko Becker, Silke Lassmann, Martin Werner, Melanie Börries, Justus Duyster, Dieter H. Heiland, Roman Sankowski, Florian Scherer","doi":"10.1038/s41698-024-00674-y","DOIUrl":"10.1038/s41698-024-00674-y","url":null,"abstract":"Despite major advances in molecular profiling and classification of primary brain tumors, personalized treatment remains limited for most patients. Here, we explored the feasibility of individual molecular profiling and the efficacy of biomarker-guided therapy for adult patients with primary brain cancers in the real-world setting within the molecular tumor board Freiburg, Germany. We analyzed genetic profiles, personalized treatment recommendations, and clinical outcomes of 102 patients with 21 brain tumor types. Alterations in the cell cycle, BRAF, and mTOR pathways most frequently led to personalized treatment recommendations. Molecularly informed therapies were recommended in 71% and implemented in 32% of patients with completed molecular diagnostics. The disease control rate following targeted treatment was 50% and the overall response rate was 30%, with a progression-free survival 2/1 ratio of at least 1.3 in 31% of patients. This study highlights the efficacy of molecularly guided treatment and the need for biomarker-stratified trials in brain cancers.","PeriodicalId":19433,"journal":{"name":"NPJ Precision Oncology","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11324882/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141982886","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
The current landscape of spatial biomarkers for prediction of response to immune checkpoint inhibition 用于预测免疫检查点抑制反应的空间生物标志物的现状。
IF 6.8 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-13 DOI: 10.1038/s41698-024-00671-1
Hannah L. Williams, Ana Leni Frei, Thibaud Koessler, Martin D. Berger, Heather Dawson, Olivier Michielin, Inti Zlobec
Enabling the examination of cell-cell relationships in tissue, spatially resolved omics technologies have revolutionised our perspectives on cancer biology. Clinically, the development of immune checkpoint inhibitors (ICI) has advanced cancer therapeutics. However, a major challenge of effective implementation is the identification of predictive biomarkers of response. In this review we examine the potential added predictive value of spatial biomarkers of response to ICI beyond current clinical benchmarks.
空间分辨全息技术能够检查组织中的细胞-细胞关系,彻底改变了我们对癌症生物学的看法。在临床上,免疫检查点抑制剂(ICI)的开发推动了癌症疗法的发展。然而,有效实施的一个主要挑战是确定反应的预测性生物标志物。在这篇综述中,我们研究了 ICI 反应空间生物标志物在当前临床基准之外的潜在附加预测价值。
{"title":"The current landscape of spatial biomarkers for prediction of response to immune checkpoint inhibition","authors":"Hannah L. Williams, Ana Leni Frei, Thibaud Koessler, Martin D. Berger, Heather Dawson, Olivier Michielin, Inti Zlobec","doi":"10.1038/s41698-024-00671-1","DOIUrl":"10.1038/s41698-024-00671-1","url":null,"abstract":"Enabling the examination of cell-cell relationships in tissue, spatially resolved omics technologies have revolutionised our perspectives on cancer biology. Clinically, the development of immune checkpoint inhibitors (ICI) has advanced cancer therapeutics. However, a major challenge of effective implementation is the identification of predictive biomarkers of response. In this review we examine the potential added predictive value of spatial biomarkers of response to ICI beyond current clinical benchmarks.","PeriodicalId":19433,"journal":{"name":"NPJ Precision Oncology","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41698-024-00671-1.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141976265","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
期刊
NPJ Precision Oncology
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