首页 > 最新文献

Seminars in oncology最新文献

英文 中文
Artificial intelligence in gynecologic oncology: A cautionary look in the Indian context 妇科肿瘤学中的人工智能:印度背景下的警示
IF 2.5 3区 医学 Q2 ONCOLOGY Pub Date : 2025-12-13 DOI: 10.1016/j.seminoncol.2025.152456
Sarita Kumari , Rudrika Chandra , Abhinav Tiwari
Artificial intelligence (AI) is making remarkable strides in the field of oncology. The potential is humongous, but the perils are understated. From the perspective of gynecologic oncologists from India, we urge everyone to take a cautionary look at the rapid AI evolution in oncology.
人工智能(AI)在肿瘤领域取得了令人瞩目的进展。潜力巨大,但风险被低估了。从印度妇科肿瘤学家的角度来看,我们敦促大家对肿瘤学中AI的快速发展持谨慎态度。
{"title":"Artificial intelligence in gynecologic oncology: A cautionary look in the Indian context","authors":"Sarita Kumari ,&nbsp;Rudrika Chandra ,&nbsp;Abhinav Tiwari","doi":"10.1016/j.seminoncol.2025.152456","DOIUrl":"10.1016/j.seminoncol.2025.152456","url":null,"abstract":"<div><div>Artificial intelligence (AI) is making remarkable strides in the field of oncology. The potential is humongous, but the perils are understated. From the perspective of gynecologic oncologists from India, we urge everyone to take a cautionary look at the rapid AI evolution in oncology.</div></div>","PeriodicalId":21750,"journal":{"name":"Seminars in oncology","volume":"53 2","pages":"Article 152456"},"PeriodicalIF":2.5,"publicationDate":"2025-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145898150","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Immuno-oncology approaches to overcome T cell exhaustion in melanoma 免疫肿瘤学方法克服黑色素瘤中的T细胞衰竭
IF 2.5 3区 医学 Q2 ONCOLOGY Pub Date : 2025-12-02 DOI: 10.1016/j.seminoncol.2025.152454
Amr Ali Mohamed Abdelgawwad El-Sehrawy , Mohammad Y Alshahrani , Muzdalifa Mejbel Fedwi , Subbulakshmi Ganesan , Zahraa Abbas Al-Khafaji , Vimal Arora , Amrita Pal , Priya Priyadarshini Nayak , Sarvar Islomov , Chou-Yi Hsu
Melanoma, a highly aggressive type of skin cancer, has undergone incredible developments in immunotherapy, particularly in modulating T-cell immunity. T cells are essential components of the antitumor immune response and can undoubtedly influence the effectiveness of melanoma treatment. This review will evaluate the roles of the different T cell subsets (CD8+, CD4+, and Tregs) in melanoma immunity. CD8+ T cells are important effectors, as they primarily recognize and kill tumor cells. However, CD8+ T cells are often dysfunctional due to exhaustion driven by chronic antigen exposure and dysfunctional immune checkpoint pathways, specifically PD-1 and CTLA-4. On the other hand, CD4+ T cells, also known as T helper cells, play a crucial role in coordinating both pro- and antitumor immune responses. In contrast to T cells, Tregs, which are often present in the tumor microenvironment, lead to immune suppression through their activity, limiting T cell activity. This review will also examine the mechanisms of T-cell exhaustion, metabolic reprogramming within the tumor microenvironment (TME) of T-cell subsets, and the role of immune checkpoint pathways, such as CTLA-4 and PD-1, in T-cell immunity. Adoptive cell therapies (ACT), specifically Tumor-Infiltrating Lymphocyte (TIL) therapy and Chimeric Antigen Receptor (CAR) T-cell therapy, have shown the ability to rejuvenate T-cells to enhance clinical outcomes. However, several resistance mechanisms and the suppressive TME presents difficulties. Future efforts will focus on combination therapies, metabolic interventions, and novel engineering techniques to overcome barriers to T-cell function exhaustion and T-cell persistence. Evaluating biomarkers associated with early prediction for therapeutic benefit and associated toxicity is important for personalizing a particular treatment. Ultimately, this review highlights the potential of targeting T-cell exhaustion to enhance the effectiveness of T-cell-based therapies in improving outcomes for melanoma patients.
黑色素瘤是一种高度侵袭性的皮肤癌,在免疫治疗方面取得了令人难以置信的进展,特别是在调节t细胞免疫方面。T细胞是抗肿瘤免疫反应的重要组成部分,无疑会影响黑色素瘤治疗的有效性。本综述将评估不同T细胞亚群(CD8+、CD4+和treg)在黑色素瘤免疫中的作用。CD8+ T细胞是重要的效应细胞,它们主要识别和杀死肿瘤细胞。然而,由于慢性抗原暴露和功能失调的免疫检查点通路(特别是PD-1和CTLA-4)驱动的衰竭,CD8+ T细胞经常功能失调。另一方面,CD4+ T细胞,也被称为T辅助细胞,在协调促肿瘤和抗肿瘤免疫反应中起着至关重要的作用。与T细胞相反,Tregs通常存在于肿瘤微环境中,通过其活性导致免疫抑制,限制T细胞活性。本综述还将研究t细胞衰竭的机制,t细胞亚群肿瘤微环境(TME)内的代谢重编程,以及免疫检查点通路(如CTLA-4和PD-1)在t细胞免疫中的作用。过继细胞疗法(ACT),特别是肿瘤浸润淋巴细胞(TIL)疗法和嵌合抗原受体(CAR) t细胞疗法,已经显示出使t细胞恢复活力以提高临床结果的能力。然而,几种耐药机制和抑制TME存在困难。未来的努力将集中在联合治疗、代谢干预和新的工程技术上,以克服t细胞功能衰竭和t细胞持久性的障碍。评估与早期预测治疗益处和相关毒性相关的生物标志物对于个性化特定治疗非常重要。最后,本综述强调了靶向t细胞衰竭的潜力,以提高t细胞为基础的治疗在改善黑色素瘤患者预后方面的有效性。
{"title":"Immuno-oncology approaches to overcome T cell exhaustion in melanoma","authors":"Amr Ali Mohamed Abdelgawwad El-Sehrawy ,&nbsp;Mohammad Y Alshahrani ,&nbsp;Muzdalifa Mejbel Fedwi ,&nbsp;Subbulakshmi Ganesan ,&nbsp;Zahraa Abbas Al-Khafaji ,&nbsp;Vimal Arora ,&nbsp;Amrita Pal ,&nbsp;Priya Priyadarshini Nayak ,&nbsp;Sarvar Islomov ,&nbsp;Chou-Yi Hsu","doi":"10.1016/j.seminoncol.2025.152454","DOIUrl":"10.1016/j.seminoncol.2025.152454","url":null,"abstract":"<div><div>Melanoma, a highly aggressive type of skin cancer, has undergone incredible developments in immunotherapy, particularly in modulating T-cell immunity. T cells are essential components of the antitumor immune response and can undoubtedly influence the effectiveness of melanoma treatment. This review will evaluate the roles of the different T cell subsets (CD8<sup>+</sup>, CD4<sup>+</sup>, and Tregs) in melanoma immunity. CD8<sup>+</sup> T cells are important effectors, as they primarily recognize and kill tumor cells. However, CD8+ T cells are often dysfunctional due to exhaustion driven by chronic antigen exposure and dysfunctional immune checkpoint pathways, specifically PD-1 and CTLA-4. On the other hand, CD4<sup>+</sup> T cells, also known as T helper cells, play a crucial role in coordinating both pro- and antitumor immune responses. In contrast to T cells, Tregs, which are often present in the tumor microenvironment, lead to immune suppression through their activity, limiting T cell activity. This review will also examine the mechanisms of T-cell exhaustion, metabolic reprogramming within the tumor microenvironment (TME) of T-cell subsets, and the role of immune checkpoint pathways, such as CTLA-4 and PD-1, in T-cell immunity. Adoptive cell therapies (ACT), specifically Tumor-Infiltrating Lymphocyte (TIL) therapy and Chimeric Antigen Receptor (CAR) T-cell therapy, have shown the ability to rejuvenate T-cells to enhance clinical outcomes. However, several resistance mechanisms and the suppressive TME presents difficulties. Future efforts will focus on combination therapies, metabolic interventions, and novel engineering techniques to overcome barriers to T-cell function exhaustion and T-cell persistence. Evaluating biomarkers associated with early prediction for therapeutic benefit and associated toxicity is important for personalizing a particular treatment. Ultimately, this review highlights the potential of targeting T-cell exhaustion to enhance the effectiveness of T-cell-based therapies in improving outcomes for melanoma patients.</div></div>","PeriodicalId":21750,"journal":{"name":"Seminars in oncology","volume":"53 1","pages":"Article 152454"},"PeriodicalIF":2.5,"publicationDate":"2025-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145839637","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting TRP channels in oral cancer: Mechanistic potential and therapeutic promise 口腔癌靶向TRP通道:机制潜力和治疗前景。
IF 2.5 3区 医学 Q2 ONCOLOGY Pub Date : 2025-11-12 DOI: 10.1016/j.seminoncol.2025.152440
Kaviyarasi Renu , Vishnu Priya Veeraraghavan , Harishkumar Madhyastha
Oral cavity cancer remains a major clinical challenge due to its aggressive nature, rapid lymphatic spread, and limited therapeutic options. Despite advancements in diagnosis and treatment, patient prognosis continues to be poor, highlighting the urgent need for novel molecular targets. This review systematically examines the critical role of calcium (Ca²⁺) ion channels, particularly transient receptor potential (TRP) channels, in the initiation and progression of oral cancer. Comprehensive searches were conducted in Scopus, Embase, Web of Science, and Google Scholar databases up to January 2025. The focus centers on TRPA1, TRPV1-4, TRPM2, TRPM6, and TRPM8 channels, emphasizing their dysregulated expression, altered functionality, and downstream signaling in oral squamous cell carcinoma (OSCC). Emerging evidence demonstrates that aberrant TRP channel activity contributes to enhanced cell proliferation, migration, invasion, and survival, thereby promoting oral carcinogenesis. Additionally, the review explores the interplay between TRP-mediated calcium signaling and oncogenic pathways such as PI3K/AKT and MAPK, elucidating their collective impact on tumor behavior. By integrating insights from molecular biology, pharmacology, and clinical studies, this work underscores the therapeutic potential of targeting TRP channels as a novel approach in oral cancer management. Future research directions include delineating channel-protein interactions and developing selective TRP inhibitors to improve treatment outcomes.
口腔癌由于其侵袭性、快速淋巴扩散和有限的治疗选择,仍然是一个主要的临床挑战。尽管诊断和治疗取得了进步,但患者预后仍然很差,迫切需要新的分子靶点。这篇综述系统地研究了钙(Ca 2 +)离子通道,特别是瞬时受体电位(TRP)通道在口腔癌的发生和发展中的关键作用。在Scopus、Embase、Web of Science和谷歌Scholar数据库中进行了截至2025年1月的综合检索。重点关注TRPA1、TRPV1-4、TRPM2、TRPM6和TRPM8通道,强调它们在口腔鳞状细胞癌(OSCC)中的表达失调、功能改变和下游信号传导。越来越多的证据表明,异常的TRP通道活性有助于增强细胞的增殖、迁移、侵袭和存活,从而促进口腔癌的发生。此外,本综述探讨了trp介导的钙信号与PI3K/AKT和MAPK等致癌途径之间的相互作用,阐明了它们对肿瘤行为的共同影响。通过整合分子生物学、药理学和临床研究的见解,这项工作强调了靶向TRP通道作为口腔癌治疗新方法的治疗潜力。未来的研究方向包括描述通道-蛋白质相互作用和开发选择性TRP抑制剂以改善治疗效果。
{"title":"Targeting TRP channels in oral cancer: Mechanistic potential and therapeutic promise","authors":"Kaviyarasi Renu ,&nbsp;Vishnu Priya Veeraraghavan ,&nbsp;Harishkumar Madhyastha","doi":"10.1016/j.seminoncol.2025.152440","DOIUrl":"10.1016/j.seminoncol.2025.152440","url":null,"abstract":"<div><div>Oral cavity cancer remains a major clinical challenge due to its aggressive nature, rapid lymphatic spread, and limited therapeutic options. Despite advancements in diagnosis and treatment, patient prognosis continues to be poor, highlighting the urgent need for novel molecular targets. This review systematically examines the critical role of calcium (Ca²⁺) ion channels, particularly transient receptor potential (TRP) channels, in the initiation and progression of oral cancer. Comprehensive searches were conducted in Scopus, Embase, Web of Science, and Google Scholar databases up to January 2025. The focus centers on TRPA1, TRPV1-4, TRPM2, TRPM6, and TRPM8 channels, emphasizing their dysregulated expression, altered functionality, and downstream signaling in oral squamous cell carcinoma (OSCC). Emerging evidence demonstrates that aberrant TRP channel activity contributes to enhanced cell proliferation, migration, invasion, and survival, thereby promoting oral carcinogenesis. Additionally, the review explores the interplay between TRP-mediated calcium signaling and oncogenic pathways such as PI3K/AKT and MAPK, elucidating their collective impact on tumor behavior. By integrating insights from molecular biology, pharmacology, and clinical studies, this work underscores the therapeutic potential of targeting TRP channels as a novel approach in oral cancer management. Future research directions include delineating channel-protein interactions and developing selective TRP inhibitors to improve treatment outcomes.</div></div>","PeriodicalId":21750,"journal":{"name":"Seminars in oncology","volume":"53 1","pages":"Article 152440"},"PeriodicalIF":2.5,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145661101","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Artificial intelligence-driven intelligent nanocarriers for cancer theranostics: A paradigm shift with focus on brain tumors 用于癌症治疗的人工智能驱动的智能纳米载体:以脑肿瘤为重点的范式转变。
IF 2.5 3区 医学 Q2 ONCOLOGY Pub Date : 2025-11-10 DOI: 10.1016/j.seminoncol.2025.152429
Mehrab Pourmadadi , Salar Mohammadi Shabestari , Hamidreza Abdouss , Abbas Rahdar , Sonia Fathi-Karkan , Sadanand Pandey
Artificial intelligence (AI) and nanotechnology are revolutionizing brain cancer theranostics by enhancing drug delivery and diagnostic accuracy. This review examines AI-enhanced engineering strategies for developing intelligent nanocarriers that target glioblastoma and other metastatic central nervous system malignancies. AI encompasses several computational methods, including machine learning (ML) and its subset deep learning (DL). Here, ML algorithms learn design rules for nanocarriers, and DL networks intricate pattern recognition for tumor segmentation and adaptive release. These approaches enable stimuli-responsive nanocarriers to react to tumor microenvironmental signals (eg, pH, enzyme activity) and external stimuli (eg, ultrasound), optimizing targeted medication release while minimizing off-target effects. Magnetic resonance imaging (MRI) and positron emission tomography (PET), in conjunction with AI, enhance tumor detection and segmentation, while the integration of multiomics data facilitates tailored treatment planning. Advanced technologies encompass transferrin-functionalized nanoparticles for traversing the blood-brain barrier (BBB) and dual-stimuli-responsive drug delivery systems. Notwithstanding general progress, apprehensions surrounding batch variability and industrial scalability persist. This review also addresses ethical concerns and cost disparities associated with AI-based therapeutics. The primary development target areas are federated learning for data privacy, explainable artificial intelligence (XAI) for regulatory transparency, and quantum ML for molecular-scale optimization. This paper charts the course to patient-specific, scalable neuro-oncology nanomedicine through the convergence of computational modeling, intelligent materials, and advanced imaging modalities. These themes are explored in greater detail in the introduction, where we lay the groundwork for intelligent nanocarriers, their design with the help of AI, and the clinical need for diagnostics-therapeutics convergence in brain cancer.
人工智能(AI)和纳米技术通过增强药物传递和诊断准确性,正在彻底改变脑癌治疗方法。本文综述了人工智能增强的工程策略,用于开发针对胶质母细胞瘤和其他转移性中枢神经系统恶性肿瘤的智能纳米载体。人工智能包括几种计算方法,包括机器学习(ML)及其子集深度学习(DL)。在这里,机器学习算法学习纳米载体的设计规则,深度学习网络复杂的模式识别用于肿瘤分割和自适应释放。这些方法使刺激反应性纳米载体能够对肿瘤微环境信号(如pH值、酶活性)和外部刺激(如超声)做出反应,优化靶向药物释放,同时最大限度地减少脱靶效应。磁共振成像(MRI)和正电子发射断层扫描(PET)与人工智能相结合,增强了肿瘤的检测和分割,而多组学数据的整合有助于制定量身定制的治疗计划。先进的技术包括用于穿越血脑屏障(BBB)的转铁蛋白功能化纳米颗粒和双刺激反应性药物递送系统。尽管总体上取得了进展,但对批处理可变性和工业可扩展性的担忧仍然存在。本综述还讨论了与人工智能治疗相关的伦理问题和成本差异。主要的发展目标领域是用于数据隐私的联邦学习、用于监管透明度的可解释人工智能(XAI)和用于分子尺度优化的量子机器学习。本文通过计算建模、智能材料和先进成像方式的融合,描绘了患者特异性、可扩展的神经肿瘤纳米医学的过程。这些主题在引言中进行了更详细的探讨,我们为智能纳米载体、它们在人工智能的帮助下的设计以及脑癌诊断治疗融合的临床需求奠定了基础。
{"title":"Artificial intelligence-driven intelligent nanocarriers for cancer theranostics: A paradigm shift with focus on brain tumors","authors":"Mehrab Pourmadadi ,&nbsp;Salar Mohammadi Shabestari ,&nbsp;Hamidreza Abdouss ,&nbsp;Abbas Rahdar ,&nbsp;Sonia Fathi-Karkan ,&nbsp;Sadanand Pandey","doi":"10.1016/j.seminoncol.2025.152429","DOIUrl":"10.1016/j.seminoncol.2025.152429","url":null,"abstract":"<div><div>Artificial intelligence (AI) and nanotechnology are revolutionizing brain cancer theranostics by enhancing drug delivery and diagnostic accuracy. This review examines AI-enhanced engineering strategies for developing intelligent nanocarriers that target glioblastoma and other metastatic central nervous system malignancies. AI encompasses several computational methods, including machine learning (ML) and its subset deep learning (DL). Here, ML algorithms learn design rules for nanocarriers, and DL networks intricate pattern recognition for tumor segmentation and adaptive release. These approaches enable stimuli-responsive nanocarriers to react to tumor microenvironmental signals (eg, pH, enzyme activity) and external stimuli (eg, ultrasound), optimizing targeted medication release while minimizing off-target effects. Magnetic resonance imaging (MRI) and positron emission tomography (PET), in conjunction with AI, enhance tumor detection and segmentation, while the integration of multiomics data facilitates tailored treatment planning. Advanced technologies encompass transferrin-functionalized nanoparticles for traversing the blood-brain barrier (BBB) and dual-stimuli-responsive drug delivery systems. Notwithstanding general progress, apprehensions surrounding batch variability and industrial scalability persist. This review also addresses ethical concerns and cost disparities associated with AI-based therapeutics. The primary development target areas are federated learning for data privacy, explainable artificial intelligence (XAI) for regulatory transparency, and quantum ML for molecular-scale optimization. This paper charts the course to patient-specific, scalable neuro-oncology nanomedicine through the convergence of computational modeling, intelligent materials, and advanced imaging modalities. These themes are explored in greater detail in the introduction, where we lay the groundwork for intelligent nanocarriers, their design with the help of AI, and the clinical need for diagnostics-therapeutics convergence in brain cancer.</div></div>","PeriodicalId":21750,"journal":{"name":"Seminars in oncology","volume":"52 6","pages":"Article 152429"},"PeriodicalIF":2.5,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145496608","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
UBE2T promotes papillary thyroid carcinoma progression by activating the JAK/STAT3 pathway via negative regulation of SOCS2 UBE2T通过负调控SOCS2激活JAK/STAT3通路,促进甲状腺乳头状癌的进展。
IF 2.5 3区 医学 Q2 ONCOLOGY Pub Date : 2025-11-08 DOI: 10.1016/j.seminoncol.2025.152439
Lijun Zhang , Chengyuan Li , Jianing Zhou , Xiang Zhang , Haisheng Fang , Jingsheng Cai , Houchao Tong , Jianfei Wen , Heda Zhang , Meiping Shen , Yan Si
Papillary thyroid carcinoma (PTC) exhibits aggressive behaviors such as tumor invasion and lymph node metastasis that critically influence prognosis, yet reliable predictors of invasiveness remain elusive. This study investigated the molecular mechanisms through which ubiquitin-conjugating enzyme E2T (UBE2T) drives PTC progression. Bioinformatics analysis of TCGA/GEO datasets and validation with institutional clinical samples revealed UBE2T overexpression correlated with advanced clinicopathological features. Functional experiments demonstrated that UBE2T overexpression enhanced PTC cell invasiveness, while its knockdown suppressed malignant behaviors. Mechanistically, co-immunoprecipitation identified cytokine signaling suppressor 2 (SOCS2) as a key interactor mediating UBE2T's effects on JAK-STAT3 pathway activation. Rescue experiments and immunofluorescence confirmed UBE2T promotes oncogenesis by destabilizing SOCS2, thereby relieving its inhibition of STAT3 phosphorylation. These findings establish UBE2T as a novel regulator of PTC progression through SOCS2/JAK-STAT3 axis manipulation, providing potential therapeutic targets to mitigate metastasis and recurrence in aggressive thyroid carcinomas.
甲状腺乳头状癌(PTC)表现出侵袭性行为,如肿瘤侵袭和淋巴结转移,这对预后有重要影响,但侵袭性的可靠预测指标仍然难以捉摸。本研究探讨了泛素偶联酶E2T (UBE2T)驱动PTC进展的分子机制。TCGA/GEO数据集的生物信息学分析和机构临床样本验证显示,UBE2T过表达与晚期临床病理特征相关。功能实验表明,UBE2T过表达增强了PTC细胞的侵袭性,而UBE2T敲低则抑制了PTC细胞的恶性行为。在机制上,共免疫沉淀鉴定细胞因子信号抑制因子2 (SOCS2)是介导UBE2T对JAK-STAT3通路激活作用的关键相互作用因子。救援实验和免疫荧光证实,UBE2T通过破坏SOCS2的稳定促进肿瘤发生,从而减轻其对STAT3磷酸化的抑制。这些研究结果表明,UBE2T通过SOCS2/JAK-STAT3轴调控PTC进展,为减轻侵袭性甲状腺癌的转移和复发提供了潜在的治疗靶点。
{"title":"UBE2T promotes papillary thyroid carcinoma progression by activating the JAK/STAT3 pathway via negative regulation of SOCS2","authors":"Lijun Zhang ,&nbsp;Chengyuan Li ,&nbsp;Jianing Zhou ,&nbsp;Xiang Zhang ,&nbsp;Haisheng Fang ,&nbsp;Jingsheng Cai ,&nbsp;Houchao Tong ,&nbsp;Jianfei Wen ,&nbsp;Heda Zhang ,&nbsp;Meiping Shen ,&nbsp;Yan Si","doi":"10.1016/j.seminoncol.2025.152439","DOIUrl":"10.1016/j.seminoncol.2025.152439","url":null,"abstract":"<div><div>Papillary thyroid carcinoma (PTC) exhibits aggressive behaviors such as tumor invasion and lymph node metastasis that critically influence prognosis, yet reliable predictors of invasiveness remain elusive. This study investigated the molecular mechanisms through which ubiquitin-conjugating enzyme E2T (UBE2T) drives PTC progression. Bioinformatics analysis of TCGA/GEO datasets and validation with institutional clinical samples revealed UBE2T overexpression correlated with advanced clinicopathological features. Functional experiments demonstrated that UBE2T overexpression enhanced PTC cell invasiveness, while its knockdown suppressed malignant behaviors. Mechanistically, co-immunoprecipitation identified cytokine signaling suppressor 2 (SOCS2) as a key interactor mediating UBE2T's effects on JAK-STAT3 pathway activation. Rescue experiments and immunofluorescence confirmed UBE2T promotes oncogenesis by destabilizing SOCS2, thereby relieving its inhibition of STAT3 phosphorylation. These findings establish UBE2T as a novel regulator of PTC progression through SOCS2/JAK-STAT3 axis manipulation, providing potential therapeutic targets to mitigate metastasis and recurrence in aggressive thyroid carcinomas.</div></div>","PeriodicalId":21750,"journal":{"name":"Seminars in oncology","volume":"53 1","pages":"Article 152439"},"PeriodicalIF":2.5,"publicationDate":"2025-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145661207","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Corrigendum to “Are we there yet? Gut microbiota for cancer diagnosis, prognosis and treatment” [Seminars in Oncology Volume 52, Issue 4, 2025, 152376] “Are we there yet?”肠道微生物群对癌症的诊断、预后和治疗”[肿瘤研讨会,第52卷,第4期,2025年,152376]。
IF 2.5 3区 医学 Q2 ONCOLOGY Pub Date : 2025-11-06 DOI: 10.1016/j.seminoncol.2025.152432
Carlos Ordóñez , Sara Zurita , Giuliana Ramírez , Fernanda Cordeiro , Katherine Garcia-Matamoros , Fuad Huaman-Garaicoa , Andrea Orellana-Manzano , Lorena Sandoya-Onofre , Juan Roca-Pogo , Diana Carvajal-Aldaz
{"title":"Corrigendum to “Are we there yet? Gut microbiota for cancer diagnosis, prognosis and treatment” [Seminars in Oncology Volume 52, Issue 4, 2025, 152376]","authors":"Carlos Ordóñez ,&nbsp;Sara Zurita ,&nbsp;Giuliana Ramírez ,&nbsp;Fernanda Cordeiro ,&nbsp;Katherine Garcia-Matamoros ,&nbsp;Fuad Huaman-Garaicoa ,&nbsp;Andrea Orellana-Manzano ,&nbsp;Lorena Sandoya-Onofre ,&nbsp;Juan Roca-Pogo ,&nbsp;Diana Carvajal-Aldaz","doi":"10.1016/j.seminoncol.2025.152432","DOIUrl":"10.1016/j.seminoncol.2025.152432","url":null,"abstract":"","PeriodicalId":21750,"journal":{"name":"Seminars in oncology","volume":"52 6","pages":"Article 152432"},"PeriodicalIF":2.5,"publicationDate":"2025-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145471818","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Colchicine as an anti-inflammatory agent improving cancer prognosis: A therapeutic repurposing perspective 秋水仙碱作为抗炎剂改善癌症预后:一种治疗再利用的观点
IF 2.5 3区 医学 Q2 ONCOLOGY Pub Date : 2025-11-04 DOI: 10.1016/j.seminoncol.2025.152437
Sigal Matza Porges PhD , Oded Shamriz MD, PhD , Shlomo Z Ben-Sasson PhD
In this Perspective, we highlight colchicine, a centuries-old, widely available anti-inflammatory drug, as a promising therapeutic candidate in oncology. We synthesize evidence demonstrating its ability to target central pathways of cancer biology, including chronic inflammation, angiogenesis, cytoskeletal remodeling, and autophagy, while also mitigating treatment-related comorbidities such as cardiovascular disease.
在这方面,我们强调秋水仙碱,一种具有数百年历史,广泛使用的抗炎药物,作为肿瘤治疗的有希望的候选药物。我们综合证据证明其能够靶向癌症生物学的中心通路,包括慢性炎症、血管生成、细胞骨架重塑和自噬,同时还能减轻治疗相关的合并症,如心血管疾病。
{"title":"Colchicine as an anti-inflammatory agent improving cancer prognosis: A therapeutic repurposing perspective","authors":"Sigal Matza Porges PhD ,&nbsp;Oded Shamriz MD, PhD ,&nbsp;Shlomo Z Ben-Sasson PhD","doi":"10.1016/j.seminoncol.2025.152437","DOIUrl":"10.1016/j.seminoncol.2025.152437","url":null,"abstract":"<div><div>In this Perspective, we highlight colchicine, a centuries-old, widely available anti-inflammatory drug, as a promising therapeutic candidate in oncology. We synthesize evidence demonstrating its ability to target central pathways of cancer biology, including chronic inflammation, angiogenesis, cytoskeletal remodeling, and autophagy, while also mitigating treatment-related comorbidities such as cardiovascular disease.</div></div>","PeriodicalId":21750,"journal":{"name":"Seminars in oncology","volume":"53 1","pages":"Article 152437"},"PeriodicalIF":2.5,"publicationDate":"2025-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145568831","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Genome editing of immune checkpoints: CRISPR-mediated PD-1 inhibition in cancer 免疫检查点的基因组编辑:crispr介导的PD-1在癌症中的抑制作用
IF 2.5 3区 医学 Q2 ONCOLOGY Pub Date : 2025-11-04 DOI: 10.1016/j.seminoncol.2025.152438
SuleimanIbrahim Mohammad , A.K. Kareem , Asokan Vasudevan , MM Rekha , Majid S. Jabir , PriyaPriyadarshini Nayak , Zahraa AlKhafaje , Vimal Arora , WesamR Kadhum , Kattela Chennakesavulu
The programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) immune checkpoint is a primary mechanism by which tumors evade immune surveillance, limiting the efficacy of cytotoxic T lymphocytes (CTLs) and tumor-infiltrating lymphocytes (TILs). Although immune checkpoint blockade therapies have revolutionized cancer treatment, their efficacy is restricted by acquired resistance, T-cell exhaustion, and tumor heterogeneity. The advent of CRISPR-Cas9 genome editing provides a precise and versatile approach to disrupt PD-1 or PD-L1, directly enhancing anti-tumor immune responses. Preclinical studies demonstrate that ex vivo PD-1 knockout in primary human T cells or TILs enhances proliferation, cytokine production, and cytotoxicity, resulting in improved tumor clearance in xenograft and humanized mouse models. In chimeric antigen receptor (CAR) T cell therapy, CRISPR-mediated disruption of PD-1 improves effector function, persistence, and resistance to exhaustion, with universal and allogeneic CAR-T platforms benefiting from multiplex genome editing. Direct PD-L1 knockout in tumor cells, often facilitated via nanoparticle- or biomaterial-assisted delivery, reshapes the immunosuppressive tumor microenvironment, promotes T cell infiltration, and enhances the efficacy of adoptive cellular therapy. Combination approaches integrating PD-1 editing with viral antigen targeting, long noncoding RNA (lncRNA) modulation, or conventional checkpoint blockade demonstrate synergistic anti-tumor effects. Clinically, early-phase trials in non-small cell lung cancer, mesothelin-positive solid tumors, and hematological malignancies establish the feasibility, safety, and preliminary efficacy of PD-1-deficient T cells. Despite these promising outcomes, challenges such as off-target effects, delivery efficiency, immunogenicity, long-term persistence, and regulatory considerations remain. This review aims to comprehensively evaluate preclinical and clinical studies investigating CRISPR-mediated PD-1/PD-L1 inhibition across various cancers, summarize mechanistic insights, and highlight translational opportunities and challenges for clinical implementation.
程序性细胞死亡蛋白1 (PD-1)/程序性死亡配体1 (PD-L1)免疫检查点是肿瘤逃避免疫监视的主要机制,限制了细胞毒性T淋巴细胞(ctl)和肿瘤浸润淋巴细胞(TILs)的功效。尽管免疫检查点阻断疗法已经彻底改变了癌症治疗,但其疗效受到获得性耐药、t细胞衰竭和肿瘤异质性的限制。CRISPR-Cas9基因组编辑的出现提供了一种精确和通用的方法来破坏PD-1或PD-L1,直接增强抗肿瘤免疫反应。临床前研究表明,在原代人T细胞或TILs中体外敲除PD-1可增强增殖、细胞因子产生和细胞毒性,从而提高异种移植和人源化小鼠模型的肿瘤清除率。在嵌合抗原受体(CAR) T细胞治疗中,crispr介导的PD-1破坏改善了效应器功能、持久性和抗衰竭性,通用和异体CAR-T平台受益于多重基因组编辑。在肿瘤细胞中直接敲除PD-L1,通常通过纳米颗粒或生物材料辅助递送,重塑免疫抑制的肿瘤微环境,促进T细胞浸润,并增强过继细胞治疗的疗效。将PD-1编辑与病毒抗原靶向、长链非编码RNA (lncRNA)调节或传统检查点阻断相结合的联合方法显示出协同抗肿瘤作用。临床上,在非小细胞肺癌、间皮素阳性实体瘤和血液系统恶性肿瘤中的早期试验证实了pd -1缺陷T细胞的可行性、安全性和初步疗效。尽管取得了这些有希望的结果,但诸如脱靶效应、给药效率、免疫原性、长期持久性和监管考虑等挑战仍然存在。本综述旨在全面评估研究crispr介导的PD-1/PD-L1抑制在各种癌症中的临床前和临床研究,总结机制见解,并强调临床实施的转化机遇和挑战。
{"title":"Genome editing of immune checkpoints: CRISPR-mediated PD-1 inhibition in cancer","authors":"SuleimanIbrahim Mohammad ,&nbsp;A.K. Kareem ,&nbsp;Asokan Vasudevan ,&nbsp;MM Rekha ,&nbsp;Majid S. Jabir ,&nbsp;PriyaPriyadarshini Nayak ,&nbsp;Zahraa AlKhafaje ,&nbsp;Vimal Arora ,&nbsp;WesamR Kadhum ,&nbsp;Kattela Chennakesavulu","doi":"10.1016/j.seminoncol.2025.152438","DOIUrl":"10.1016/j.seminoncol.2025.152438","url":null,"abstract":"<div><div>The programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) immune checkpoint is a primary mechanism by which tumors evade immune surveillance, limiting the efficacy of cytotoxic T lymphocytes (CTLs) and tumor-infiltrating lymphocytes (TILs). Although immune checkpoint blockade therapies have revolutionized cancer treatment, their efficacy is restricted by acquired resistance, T-cell exhaustion, and tumor heterogeneity. The advent of CRISPR-Cas9 genome editing provides a precise and versatile approach to disrupt PD-1 or PD-L1, directly enhancing anti-tumor immune responses. Preclinical studies demonstrate that ex vivo PD-1 knockout in primary human T cells or TILs enhances proliferation, cytokine production, and cytotoxicity, resulting in improved tumor clearance in xenograft and humanized mouse models. In chimeric antigen receptor (CAR) T cell therapy, CRISPR-mediated disruption of PD-1 improves effector function, persistence, and resistance to exhaustion, with universal and allogeneic CAR-T platforms benefiting from multiplex genome editing. Direct PD-L1 knockout in tumor cells, often facilitated via nanoparticle- or biomaterial-assisted delivery, reshapes the immunosuppressive tumor microenvironment, promotes T cell infiltration, and enhances the efficacy of adoptive cellular therapy. Combination approaches integrating PD-1 editing with viral antigen targeting, long noncoding RNA (lncRNA) modulation, or conventional checkpoint blockade demonstrate synergistic anti-tumor effects. Clinically, early-phase trials in non-small cell lung cancer, mesothelin-positive solid tumors, and hematological malignancies establish the feasibility, safety, and preliminary efficacy of PD-1-deficient T cells. Despite these promising outcomes, challenges such as off-target effects, delivery efficiency, immunogenicity, long-term persistence, and regulatory considerations remain. This review aims to comprehensively evaluate preclinical and clinical studies investigating CRISPR-mediated PD-1/PD-L1 inhibition across various cancers, summarize mechanistic insights, and highlight translational opportunities and challenges for clinical implementation.</div></div>","PeriodicalId":21750,"journal":{"name":"Seminars in oncology","volume":"53 1","pages":"Article 152438"},"PeriodicalIF":2.5,"publicationDate":"2025-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145614817","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CAR-T cell therapy: A therapeutic strategy for cancer treatment CAR-T细胞疗法:癌症治疗的一种治疗策略。
IF 2.5 3区 医学 Q2 ONCOLOGY Pub Date : 2025-11-03 DOI: 10.1016/j.seminoncol.2025.152430
Shabana Sharif, Upma Sharma, Ashok Kumar Yadav
In the twenty-first century, chimeric antigen receptor (CAR)-T cell therapy has transformed cancer immunotherapy by offering novel approaches and life-saving treatments for illnesses that were previously incurable. This method is currently being used in clinical trials for solid tumors like prostate cancer and glioblastoma, as well as viral and autoimmune illnesses. It has demonstrated impressive efficacy in treating a variety of hematological malignancies. Harvesting a patient's T cells, genetically modifying them using viral vectors to express CARs that target specific antigens, and then reintroducing the altered cells into the patient is the process of CAR-T cell therapy. These CAR-T cells detect and destroy target cells specifically, regardless of the presence of the major histocompatibility complex (MHC) antigen. The major turning points in the development of CAR-T cells, from their creation to their use in medicine, are highlighted in this overview. It describes how CAR-T cells were developed historically, highlights the significant advancements that have made them a ground-breaking treatment, and talks about the obstacles that still need to be overcome, such as the high cost of production, restricted availability, and toxicity problems like cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. The review also looks at the field's future developments with the goals of increasing therapeutic uses, minimizing toxicity, and maximizing efficacy. With safer and more efficient CAR T cell therapies being developed, we are optimistic that a larger group of cancer patients may soon benefit from this innovative treatment.
在21世纪,嵌合抗原受体(CAR)-T细胞疗法通过为以前无法治愈的疾病提供新的方法和挽救生命的治疗方法,改变了癌症免疫疗法。这种方法目前被用于前列腺癌和胶质母细胞瘤等实体肿瘤的临床试验,以及病毒和自身免疫性疾病。它在治疗多种血液系统恶性肿瘤方面表现出令人印象深刻的疗效。获取患者的T细胞,用病毒载体对其进行基因修饰,使其表达针对特定抗原的car,然后将改变后的细胞重新引入患者体内,这就是CAR-T细胞疗法的过程。不管主要组织相容性复合体(MHC)抗原是否存在,这些CAR-T细胞特异性地检测和破坏靶细胞。本文概述了CAR-T细胞发展的主要转折点,从它们的产生到它们在医学上的应用。它描述了CAR-T细胞的历史发展过程,强调了使其成为突破性治疗方法的重大进展,并讨论了仍然需要克服的障碍,例如生产成本高,可用性有限,以及细胞因子释放综合征和免疫效应细胞相关神经毒性综合征等毒性问题。该综述还着眼于该领域的未来发展,目标是增加治疗用途,最小化毒性和最大化功效。随着更安全、更有效的CAR - T细胞疗法的开发,我们乐观地认为,更多的癌症患者可能很快就会从这种创新疗法中受益。
{"title":"CAR-T cell therapy: A therapeutic strategy for cancer treatment","authors":"Shabana Sharif,&nbsp;Upma Sharma,&nbsp;Ashok Kumar Yadav","doi":"10.1016/j.seminoncol.2025.152430","DOIUrl":"10.1016/j.seminoncol.2025.152430","url":null,"abstract":"<div><div>In the twenty-first century, chimeric antigen receptor (CAR)-T cell therapy has transformed cancer immunotherapy by offering novel approaches and life-saving treatments for illnesses that were previously incurable. This method is currently being used in clinical trials for solid tumors like prostate cancer and glioblastoma, as well as viral and autoimmune illnesses. It has demonstrated impressive efficacy in treating a variety of hematological malignancies. Harvesting a patient's T cells, genetically modifying them using viral vectors to express CARs that target specific antigens, and then reintroducing the altered cells into the patient is the process of CAR-T cell therapy. These CAR-T cells detect and destroy target cells specifically, regardless of the presence of the major histocompatibility complex (MHC) antigen. The major turning points in the development of CAR-T cells, from their creation to their use in medicine, are highlighted in this overview. It describes how CAR-T cells were developed historically, highlights the significant advancements that have made them a ground-breaking treatment, and talks about the obstacles that still need to be overcome, such as the high cost of production, restricted availability, and toxicity problems like cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. The review also looks at the field's future developments with the goals of increasing therapeutic uses, minimizing toxicity, and maximizing efficacy. With safer and more efficient CAR T cell therapies being developed, we are optimistic that a larger group of cancer patients may soon benefit from this innovative treatment.</div></div>","PeriodicalId":21750,"journal":{"name":"Seminars in oncology","volume":"52 6","pages":"Article 152430"},"PeriodicalIF":2.5,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145445690","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pretreatment eosinophilia as a biomarker for adverse outcomes in non-small cell lung cancer patients receiving immune checkpoint inhibitors: A systematic review and meta-analysis 预处理嗜酸性粒细胞增多作为接受免疫检查点抑制剂的非小细胞肺癌患者不良结局的生物标志物:一项系统综述和荟萃分析
IF 2.5 3区 医学 Q2 ONCOLOGY Pub Date : 2025-11-03 DOI: 10.1016/j.seminoncol.2025.152431
Nency Ganatra , Jacob Thompson , Rupak Desai , Jinish Doshi , Pragya Jain , Diksha Sanjana Pasnoor , Akhil Jain
In recent years, immune checkpoint inhibitors (ICIs) have transformed the therapeutic landscape of non-small cell lung cancer (NSCLC), yet treatment response and adverse events vary widely among patients. In response, the identification of reliable pretreatment biomarkers has become a major goal for many clinicians to enhance prognostication and personalized care. As such, this systematic review and meta-analysis aimed to evaluate whether pretreatment eosinophilia is associated with adverse clinical outcomes in NSCLC patients receiving ICI therapy. Following PRISMA guidelines, a comprehensive literature search was conducted across online databases through February 2025. Eligible studies included observational designs reporting associations between baseline eosinophil levels and overall survival, progression-free survival, or immune-related adverse events (irAEs) in ICI-treated NSCLC patients. Pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated using random-effects models for both unadjusted and adjusted data. Eleven studies met inclusion criteria. Pretreatment eosinophilia was associated with a nonsignificant reduction in overall survival based on both unadjusted analyses (OR: 0.79, 95% CI: 0.42–1.51 and OR: 0.74, 95% CI: 0.53–1.03, respectively). Similarly, a nonsignificant reduction in progression-free survival was found in unadjusted models (OR: 0.78, 95% CI: 0.54–1.13), whereas adjusted data revealed a significant negative association (OR: 0.68, 95% CI: 0.58–0.80). In contrast, eosinophilia was significantly associated with increased odds of irAEs in both unadjusted and adjusted analyses (OR: 3.19, 95% CI: 2.11–4.83 and OR: 3.35, 95% CI: 2.25–5.02, respectively). These findings indicate that pretreatment eosinophilia may serve as a useful prognostic biomarker indicating increased susceptibility to irAEs and potentially poorer survival outcomes in ICI-treated NSCLC patients.
近年来,免疫检查点抑制剂(ICIs)已经改变了非小细胞肺癌(NSCLC)的治疗前景,但不同患者的治疗反应和不良事件差异很大。因此,确定可靠的预处理生物标志物已成为许多临床医生提高预后和个性化护理的主要目标。因此,本系统综述和荟萃分析旨在评估在接受ICI治疗的非小细胞肺癌患者中,预处理嗜酸性粒细胞增多是否与不良临床结果相关。按照PRISMA的指导方针,在2025年2月之前对在线数据库进行了全面的文献检索。符合条件的研究包括观察性设计,报告基线嗜酸性粒细胞水平与ici治疗的非小细胞肺癌患者的总生存期、无进展生存期或免疫相关不良事件(irAEs)之间的关联。使用随机效应模型计算未调整和调整数据的合并优势比(ORs)和95%置信区间(ci)。11项研究符合纳入标准。未经调整的两项分析显示,预处理嗜酸性粒细胞增多与总生存期无显著降低相关(OR分别为0.79,95% CI: 0.42-1.51和OR: 0.74, 95% CI: 0.53-1.03)。同样,在未调整的模型中发现无进展生存期无显著降低(OR: 0.78, 95% CI: 0.54-1.13),而调整后的数据显示显著负相关(OR: 0.68, 95% CI: 0.58-0.80)。相比之下,在未调整和调整的分析中,嗜酸性粒细胞增多与irae的发生率增加显著相关(OR: 3.19, 95% CI: 2.11-4.83; OR: 3.35, 95% CI: 2.25-5.02)。这些发现表明,预处理嗜酸性粒细胞增多可能是一种有用的预后生物标志物,表明ici治疗的非小细胞肺癌患者对irae的易感性增加,生存结果可能更差。
{"title":"Pretreatment eosinophilia as a biomarker for adverse outcomes in non-small cell lung cancer patients receiving immune checkpoint inhibitors: A systematic review and meta-analysis","authors":"Nency Ganatra ,&nbsp;Jacob Thompson ,&nbsp;Rupak Desai ,&nbsp;Jinish Doshi ,&nbsp;Pragya Jain ,&nbsp;Diksha Sanjana Pasnoor ,&nbsp;Akhil Jain","doi":"10.1016/j.seminoncol.2025.152431","DOIUrl":"10.1016/j.seminoncol.2025.152431","url":null,"abstract":"<div><div>In recent years, immune checkpoint inhibitors (ICIs) have transformed the therapeutic landscape of non-small cell lung cancer (NSCLC), yet treatment response and adverse events vary widely among patients. In response, the identification of reliable pretreatment biomarkers has become a major goal for many clinicians to enhance prognostication and personalized care. As such, this systematic review and meta-analysis aimed to evaluate whether pretreatment eosinophilia is associated with adverse clinical outcomes in NSCLC patients receiving ICI therapy. Following PRISMA guidelines, a comprehensive literature search was conducted across online databases through February 2025. Eligible studies included observational designs reporting associations between baseline eosinophil levels and overall survival, progression-free survival, or immune-related adverse events (irAEs) in ICI-treated NSCLC patients. Pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated using random-effects models for both unadjusted and adjusted data. Eleven studies met inclusion criteria. Pretreatment eosinophilia was associated with a nonsignificant reduction in overall survival based on both unadjusted analyses (OR: 0.79, 95% CI: 0.42–1.51 and OR: 0.74, 95% CI: 0.53–1.03, respectively). Similarly, a nonsignificant reduction in progression-free survival was found in unadjusted models (OR: 0.78, 95% CI: 0.54–1.13), whereas adjusted data revealed a significant negative association (OR: 0.68, 95% CI: 0.58–0.80). In contrast, eosinophilia was significantly associated with increased odds of irAEs in both unadjusted and adjusted analyses (OR: 3.19, 95% CI: 2.11–4.83 and OR: 3.35, 95% CI: 2.25–5.02, respectively). These findings indicate that pretreatment eosinophilia may serve as a useful prognostic biomarker indicating increased susceptibility to irAEs and potentially poorer survival outcomes in ICI-treated NSCLC patients.</div></div>","PeriodicalId":21750,"journal":{"name":"Seminars in oncology","volume":"52 6","pages":"Article 152431"},"PeriodicalIF":2.5,"publicationDate":"2025-11-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145445725","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Seminars in 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:604180095
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