首页 > 最新文献

Biochimica et biophysica acta. Reviews on cancer最新文献

英文 中文
Corrigendum to “Snail1 as a key prognostic biomarker of cancer-associated fibroblasts in breast tumors” [Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, volume 1880 (2025) / 189316] “Snail1作为乳腺肿瘤中癌症相关成纤维细胞的关键预后生物标志物”的修正[biochemica et Biophysica Acta (BBA) - Reviews on Cancer, volume 1880(2025) / 189316]。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-01 DOI: 10.1016/j.bbcan.2025.189462
Raúl Peña, Josep Baulida
{"title":"Corrigendum to “Snail1 as a key prognostic biomarker of cancer-associated fibroblasts in breast tumors” [Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, volume 1880 (2025) / 189316]","authors":"Raúl Peña, Josep Baulida","doi":"10.1016/j.bbcan.2025.189462","DOIUrl":"10.1016/j.bbcan.2025.189462","url":null,"abstract":"","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1880 6","pages":"Article 189462"},"PeriodicalIF":9.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145226500","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Opposing roles of YY1 and RKIP in cancer progression and therapy resistance YY1和RKIP在癌症进展和治疗耐药中的对立作用。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-01 DOI: 10.1016/j.bbcan.2025.189488
Christos Rigopoulos , Stavroula Baritaki , Ilias Georgakopoulos-Soares , Benjamin Bonavida , Apostolos Zaravinos
Yin Yang 1 (YY1) and Raf kinase inhibitory protein (RKIP, encoded by PEBP1) are multifunctional regulators with antagonistic roles in tumor biology. YY1 functions as a context-dependent transcription factor and chromatin organizer, integrating enhancer–promoter looping and super-enhancer activity to drive oncogenic transcriptional programs, immune evasion, and therapy resistance. By contrast, RKIP restrains the NF-κB, MAPK and STAT3 pathways, suppressing epithelial–mesenchymal transition, metastasis, and chemoresistance. Recent pan-cancer transcriptomic analyses highlight a recurrent pattern of YY1 upregulation and RKIP downregulation, with inverse correlations in several tumor types (e.g., lung, colorectal and kidney). Clinically, high YY1 expression is associated with poor survival and diminished responses to chemotherapy, TRAIL, and immune checkpoint blockade, whereas high RKIP predicts improved outcomes and restored therapeutic sensitivity. YY1 and RKIP also modulate key immune compartments: YY1 promotes Treg expansion, myeloid-derived suppressor cell (MDSC) accumulation, tumor-associated macrophage (TAM) polarization, and impaired antigen presentation, while RKIP counteracts these processes and enhances cytotoxic T and NK cell activity. Therapeutically, YY1 can be targeted through BET/p300 inhibitors, emerging PROTAC degraders, and RNA- or CRISPR-based approaches; RKIP restoration is under investigation via kinase and pathway modulators. Together, YY1 and RKIP constitute a regulatory axis with prognostic and predictive value, offering potential for biomarker-driven patient stratification and novel therapeutic strategies in precision oncology.
由PEBP1编码的阴阳1 (YY1)和Raf激酶抑制蛋白(RKIP)是肿瘤生物学中具有拮抗作用的多功能调节因子。YY1作为一种环境依赖的转录因子和染色质组织者,整合增强子-启动子环和超级增强子活性,以驱动致癌转录程序、免疫逃避和治疗抵抗。相比之下,RKIP抑制NF-κB、MAPK和STAT3通路,抑制上皮-间质转化、转移和化疗耐药。最近的泛癌症转录组学分析强调了YY1上调和RKIP下调的复发模式,在几种肿瘤类型(如肺癌、结直肠癌和肾癌)中呈负相关。在临床上,YY1高表达与生存率差和化疗、TRAIL和免疫检查点阻断反应减弱有关,而高RKIP预测预后改善和治疗敏感性恢复。YY1和RKIP也调节关键的免疫区室:YY1促进Treg扩增、髓源性抑制细胞(MDSC)积累、肿瘤相关巨噬细胞(TAM)极化和抗原呈递受损,而RKIP抵消这些过程并增强细胞毒性T和NK细胞活性。在治疗上,YY1可以通过BET/p300抑制剂、新兴的PROTAC降解剂和基于RNA或crispr的方法靶向治疗;RKIP恢复正在通过激酶和通路调节剂进行研究。YY1和RKIP共同构成了一个具有预后和预测价值的调控轴,为精准肿瘤学的生物标志物驱动的患者分层和新的治疗策略提供了潜力。
{"title":"Opposing roles of YY1 and RKIP in cancer progression and therapy resistance","authors":"Christos Rigopoulos ,&nbsp;Stavroula Baritaki ,&nbsp;Ilias Georgakopoulos-Soares ,&nbsp;Benjamin Bonavida ,&nbsp;Apostolos Zaravinos","doi":"10.1016/j.bbcan.2025.189488","DOIUrl":"10.1016/j.bbcan.2025.189488","url":null,"abstract":"<div><div>Yin Yang 1 (YY1) and Raf kinase inhibitory protein (RKIP, encoded by PEBP1) are multifunctional regulators with antagonistic roles in tumor biology. YY1 functions as a context-dependent transcription factor and chromatin organizer, integrating enhancer–promoter looping and super-enhancer activity to drive oncogenic transcriptional programs, immune evasion, and therapy resistance. By contrast, RKIP restrains the NF-κB, MAPK and STAT3 pathways, suppressing epithelial–mesenchymal transition, metastasis, and chemoresistance. Recent pan-cancer transcriptomic analyses highlight a recurrent pattern of YY1 upregulation and RKIP downregulation, with inverse correlations in several tumor types (e.g., lung, colorectal and kidney). Clinically, high YY1 expression is associated with poor survival and diminished responses to chemotherapy, TRAIL, and immune checkpoint blockade, whereas high RKIP predicts improved outcomes and restored therapeutic sensitivity. YY1 and RKIP also modulate key immune compartments: YY1 promotes Treg expansion, myeloid-derived suppressor cell (MDSC) accumulation, tumor-associated macrophage (TAM) polarization, and impaired antigen presentation, while RKIP counteracts these processes and enhances cytotoxic T and NK cell activity. Therapeutically, YY1 can be targeted through BET/p300 inhibitors, emerging PROTAC degraders, and RNA- or CRISPR-based approaches; RKIP restoration is under investigation via kinase and pathway modulators. Together, YY1 and RKIP constitute a regulatory axis with prognostic and predictive value, offering potential for biomarker-driven patient stratification and novel therapeutic strategies in precision oncology.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1880 6","pages":"Article 189488"},"PeriodicalIF":9.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145454487","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Nanoparticulate delivery and targeting of RNA to the brain 纳米颗粒递送和靶向RNA到大脑
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-01 DOI: 10.1016/j.bbcan.2025.189480
Aditya Gupta , Raghu Ramanathan , Chittalsinh M. Raulji , Ram I. Mahato
The blood-brain barrier (BBB) presents a critical challenge in treating central nervous system (CNS) disorders, particularly aggressive brain cancers such as glioblastoma (GBM) and medulloblastoma (MB). RNA therapies exploit endogenous cellular machinery to modulate gene expression, targeting previously undruggable pathways. RNA and CRISPR gene therapies hold transformative potential for brain cancer but demand breakthroughs for enhanced drug transport across the BBB. While clinical achievements in non-CNS diseases validate their efficacy, interdisciplinary collaboration is essential to advance nanoparticles (NPs) engineering, immune evasion, and non-invasive delivery for CNS applications. NPs are indispensable for advancing RNA therapies in brain cancer, with lipid nanoparticles (LNPs) and viral vectors leading clinical translation. Innovations in targeting (e.g., GLUT1, RVG peptide, ApoE mimetic peptide) and non-invasive delivery (e.g., focused ultrasound) are critical to overcome the BBB limitations. This review highlights the different strategies that can be utilized to deliver RNA-based therapies to the brain and summarizes the recent clinical efforts to deliver the RNA.
血脑屏障(BBB)是治疗中枢神经系统(CNS)疾病,特别是侵袭性脑癌,如胶质母细胞瘤(GBM)和髓母细胞瘤(MB)的关键挑战。RNA疗法利用内源性细胞机制来调节基因表达,靶向以前不可药物的途径。RNA和CRISPR基因疗法具有改变脑癌的潜力,但需要在增强药物跨血脑屏障运输方面取得突破。虽然在非中枢神经系统疾病中的临床成就证实了它们的疗效,但跨学科合作对于推进纳米颗粒(NPs)的工程、免疫逃避和非侵入性递送至关重要。脂质纳米颗粒(LNPs)和病毒载体在脑癌的临床转化中起着重要作用。靶向治疗(如GLUT1、RVG肽、ApoE模拟肽)和非侵入性给药(如聚焦超声)方面的创新对于克服血脑屏障的局限性至关重要。这篇综述强调了可用于将基于RNA的治疗方法传递到大脑的不同策略,并总结了最近在传递RNA方面的临床努力。
{"title":"Nanoparticulate delivery and targeting of RNA to the brain","authors":"Aditya Gupta ,&nbsp;Raghu Ramanathan ,&nbsp;Chittalsinh M. Raulji ,&nbsp;Ram I. Mahato","doi":"10.1016/j.bbcan.2025.189480","DOIUrl":"10.1016/j.bbcan.2025.189480","url":null,"abstract":"<div><div>The blood-brain barrier (BBB) presents a critical challenge in treating central nervous system (CNS) disorders, particularly aggressive brain cancers such as glioblastoma (GBM) and medulloblastoma (MB). RNA therapies exploit endogenous cellular machinery to modulate gene expression, targeting previously undruggable pathways. RNA and CRISPR gene therapies hold transformative potential for brain cancer but demand breakthroughs for enhanced drug transport across the BBB. While clinical achievements in non-CNS diseases validate their efficacy, interdisciplinary collaboration is essential to advance nanoparticles (NPs) engineering, immune evasion, and non-invasive delivery for CNS applications. NPs are indispensable for advancing RNA therapies in brain cancer, with lipid nanoparticles (LNPs) and viral vectors leading clinical translation. Innovations in targeting (e.g., GLUT1, RVG peptide, ApoE mimetic peptide) and non-invasive delivery (e.g., focused ultrasound) are critical to overcome the BBB limitations. This review highlights the different strategies that can be utilized to deliver RNA-based therapies to the brain and summarizes the recent clinical efforts to deliver the RNA.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1880 6","pages":"Article 189480"},"PeriodicalIF":9.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145412479","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Decoding the epigenetic landscape: ctDNA methylation as a game-changer in hepatocellular carcinoma management 解码表观遗传景观:ctDNA甲基化作为肝细胞癌管理的游戏规则改变者。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-01 DOI: 10.1016/j.bbcan.2025.189497
Dexin Yang , Yueru Yao , Fenfang Gui , Wuxuan Mei , Changchun Zeng
Hepatocellular carcinoma (HCC) constitutes a pressing global health issue, where conventional diagnostics like alpha-fetoprotein (AFP) and ultrasonography reveal limitations in early identification and prognostic reliability. Circulating tumor DNA (ctDNA) methylation has recently gained attention as an innovative “liquid biopsy” biomarker, offering epigenetic signatures and early disease signals through non-invasive detection. This review elucidates the mechanistic role of DNA methylation in HCC, highlighting its regulation of pivotal oncogenic cascades and its progression-dependent alterations. In addition, advanced detection techniques, such as restriction enzyme-based, enrichment-based, and bisulfite conversion-based approaches, were assessed in HCC. Moreover, the clinical applicability of ctDNA methylation in HCC was evaluated for early detection, prognostic assessment, and therapeutic surveillance. This review thoroughly outlines the current obstacles hindering the successful clinical translation of ongoing research. Furthermore, it outlines potential research directions to address these limitations and facilitate the rapid translation of findings into clinical practice. Ultimately, ctDNA methylation provides a significant advancement in deciphering the epigenetic profile of HCC, with potential benefits for precision oncology and patient outcomes.
肝细胞癌(HCC)是一个紧迫的全球健康问题,传统的诊断方法,如甲胎蛋白(AFP)和超声检查,在早期识别和预后可靠性方面存在局限性。循环肿瘤DNA (ctDNA)甲基化最近作为一种创新的“液体活检”生物标志物受到关注,通过非侵入性检测提供表观遗传特征和早期疾病信号。这篇综述阐明了DNA甲基化在HCC中的机制作用,强调了其对关键致癌级联反应及其进展依赖性改变的调节。此外,先进的检测技术,如基于限制性内切酶、基于富集和基于亚硫酸盐转化的方法,在HCC中进行了评估。此外,ctDNA甲基化在HCC早期检测、预后评估和治疗监测方面的临床适用性进行了评估。这篇综述全面概述了目前阻碍正在进行的研究成功临床转化的障碍。此外,它概述了潜在的研究方向,以解决这些限制,并促进研究结果快速转化为临床实践。最终,ctDNA甲基化在解读HCC的表观遗传谱方面提供了重大进展,对精确肿瘤学和患者预后有潜在的好处。
{"title":"Decoding the epigenetic landscape: ctDNA methylation as a game-changer in hepatocellular carcinoma management","authors":"Dexin Yang ,&nbsp;Yueru Yao ,&nbsp;Fenfang Gui ,&nbsp;Wuxuan Mei ,&nbsp;Changchun Zeng","doi":"10.1016/j.bbcan.2025.189497","DOIUrl":"10.1016/j.bbcan.2025.189497","url":null,"abstract":"<div><div>Hepatocellular carcinoma (HCC) constitutes a pressing global health issue, where conventional diagnostics like alpha-fetoprotein (AFP) and ultrasonography reveal limitations in early identification and prognostic reliability. Circulating tumor DNA (ctDNA) methylation has recently gained attention as an innovative “liquid biopsy” biomarker, offering epigenetic signatures and early disease signals through non-invasive detection. This review elucidates the mechanistic role of DNA methylation in HCC, highlighting its regulation of pivotal oncogenic cascades and its progression-dependent alterations. In addition, advanced detection techniques, such as restriction enzyme-based, enrichment-based, and bisulfite conversion-based approaches, were assessed in HCC. Moreover, the clinical applicability of ctDNA methylation in HCC was evaluated for early detection, prognostic assessment, and therapeutic surveillance. This review thoroughly outlines the current obstacles hindering the successful clinical translation of ongoing research. Furthermore, it outlines potential research directions to address these limitations and facilitate the rapid translation of findings into clinical practice. Ultimately, ctDNA methylation provides a significant advancement in deciphering the epigenetic profile of HCC, with potential benefits for precision oncology and patient outcomes.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1880 6","pages":"Article 189497"},"PeriodicalIF":9.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145544567","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The mechanics of anoikis resistance in cancer 癌症耐药机制。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-01 DOI: 10.1016/j.bbcan.2025.189498
Michela Corsini , Mattia Domenichini , Elisa Moreschi , Stefania Mitola
Metastatic cancer cells display a remarkable ability to resist the mechanical and biochemical challenges associated with detaching from the extracellular matrix and metastasize. A key adaptive mechanism in this process is resistance to anoikis. In this review, we explore the molecular and biophysical mechanisms that enable cancer cells to resist anoikis, with a focus on mechanotransduction. We discuss the roles of integrin signaling, the YAP/TAZ pathway, the mechanosensitive ion channels, and actomyosin contractility in sustaining survival under mechanical stress conditions. Furthermore, we highlight the emerging contribution of soluble mediators, particularly the myokine irisin, which acts as mechanical mimetics by activating survival pathways typically triggered by matrix engagement. We also examined how mechanical heterogeneity across tumor types and metastatic routes shapes context-specific adaptation strategies. By bridging physical forces and cell survival signaling, this review underscores mechanostransduction as fundamental driver of metastatic competence and a promising target for therapeutic intervention.
转移性癌细胞表现出非凡的能力来抵抗与细胞外基质分离和转移相关的机械和生化挑战。在这一过程中,一个关键的适应机制是对疾病的抵抗。在这篇综述中,我们探讨了使癌细胞抵抗疾病的分子和生物物理机制,重点是机械转导。我们讨论了整合素信号、YAP/TAZ通路、机械敏感离子通道和肌动球蛋白收缩性在机械应力条件下维持存活中的作用。此外,我们强调了可溶性介质的新兴贡献,特别是肌因子鸢尾素,它通过激活通常由基质接合触发的生存途径来作为机械模拟物。我们还研究了不同肿瘤类型和转移途径的机械异质性如何影响环境特异性适应策略。通过连接物理力量和细胞存活信号,本综述强调机械转导是转移能力的基本驱动因素,也是治疗干预的一个有希望的靶点。
{"title":"The mechanics of anoikis resistance in cancer","authors":"Michela Corsini ,&nbsp;Mattia Domenichini ,&nbsp;Elisa Moreschi ,&nbsp;Stefania Mitola","doi":"10.1016/j.bbcan.2025.189498","DOIUrl":"10.1016/j.bbcan.2025.189498","url":null,"abstract":"<div><div>Metastatic cancer cells display a remarkable ability to resist the mechanical and biochemical challenges associated with detaching from the extracellular matrix and metastasize. A key adaptive mechanism in this process is resistance to anoikis. In this review, we explore the molecular and biophysical mechanisms that enable cancer cells to resist anoikis, with a focus on mechanotransduction. We discuss the roles of integrin signaling, the YAP/TAZ pathway, the mechanosensitive ion channels, and actomyosin contractility in sustaining survival under mechanical stress conditions. Furthermore, we highlight the emerging contribution of soluble mediators, particularly the myokine irisin, which acts as mechanical mimetics by activating survival pathways typically triggered by matrix engagement. We also examined how mechanical heterogeneity across tumor types and metastatic routes shapes context-specific adaptation strategies. By bridging physical forces and cell survival signaling, this review underscores mechanostransduction as fundamental driver of metastatic competence and a promising target for therapeutic intervention.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1880 6","pages":"Article 189498"},"PeriodicalIF":9.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145566661","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
mRNA cancer vaccine: A novel and potential immunotherapy for multiple myeloma mRNA癌症疫苗:一种新的和潜在的多发性骨髓瘤免疫疗法。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-01 DOI: 10.1016/j.bbcan.2025.189493
Yiming Feng , Yufeng Du , Chengtao Zhang , Fang Xie , Jinsong Yan
Multiple myeloma (MM) is a malignant haematologic tumour originating from bone marrow plasma cells, characterised by the abnormal proliferation of monoclonal plasma cells, invasion of the bone marrow, and resulting end-organ damage, with continuous clinical progression. Although standardised treatment regimens and the application of novel cellular immunotherapies have helped control disease progression, the risks of drug resistance and relapse remain, and a complete cure is currently not achievable. mRNA cancer vaccines aim to target personalized neoantigens, inducing strong and specific anti-tumour immune responses, and have garnered widespread attention for their tremendous potential in cancer prevention and treatment. Compared to traditional cancer vaccine platforms, mRNA cancer vaccines offer unique advantages such as high safety, strong immunogenicity, personalized targeted immunotherapy, shorter development cycles, and lower costs, making them a revolutionary tool for achieving the goals of precision medicine. Given the unique advantages of MM, such as antigen lineage specificity, pathological mechanism and therapeutic needs alignment, clear safety profile and treatment endpoints, clinical window, and compatibility with multi-strategy combination therapies, we believe that exploring the use of mRNA cancer vaccines for the treatment of MM is both necessary and promising. In this review, we aimed to summarize the progress of immunotherapy for MM and the current application status of various antigens and vaccines. On this basis, we analysed the specificity and feasibility of mRNA cancer vaccines in MM and proposed strategies for their clinical management and optimisation. We believe that mRNA cancer vaccines have the potential to redefine the treatment paradigm for MM, offering new solutions for patients with refractory and relapsed disease.
多发性骨髓瘤(Multiple myeloma, MM)是一种起源于骨髓浆细胞的恶性血液学肿瘤,其特点是单克隆浆细胞异常增殖,侵袭骨髓,导致终末器官损害,临床进展不断。尽管标准化的治疗方案和新型细胞免疫疗法的应用有助于控制疾病的进展,但耐药和复发的风险仍然存在,目前无法实现完全治愈。mRNA肿瘤疫苗以个体化新抗原为靶点,诱导强而特异性的抗肿瘤免疫反应,在癌症预防和治疗方面具有巨大潜力,受到广泛关注。与传统的癌症疫苗平台相比,mRNA癌症疫苗具有安全性高、免疫原性强、个性化靶向免疫治疗、开发周期短、成本低等独特优势,是实现精准医疗目标的革命性工具。鉴于MM的独特优势,如抗原谱系特异性、病理机制和治疗需求一致性、明确的安全性和治疗终点、临床窗口以及与多策略联合治疗的兼容性,我们认为探索使用mRNA癌症疫苗治疗MM是必要的和有希望的。本文就MM的免疫治疗进展及各种抗原和疫苗的应用现状进行综述。在此基础上,我们分析了mRNA癌症疫苗在MM中的特异性和可行性,并提出了其临床管理和优化策略。我们相信mRNA癌症疫苗有可能重新定义MM的治疗模式,为难治性和复发性疾病患者提供新的解决方案。
{"title":"mRNA cancer vaccine: A novel and potential immunotherapy for multiple myeloma","authors":"Yiming Feng ,&nbsp;Yufeng Du ,&nbsp;Chengtao Zhang ,&nbsp;Fang Xie ,&nbsp;Jinsong Yan","doi":"10.1016/j.bbcan.2025.189493","DOIUrl":"10.1016/j.bbcan.2025.189493","url":null,"abstract":"<div><div>Multiple myeloma (MM) is a malignant haematologic tumour originating from bone marrow plasma cells, characterised by the abnormal proliferation of monoclonal plasma cells, invasion of the bone marrow, and resulting end-organ damage, with continuous clinical progression. Although standardised treatment regimens and the application of novel cellular immunotherapies have helped control disease progression, the risks of drug resistance and relapse remain, and a complete cure is currently not achievable. mRNA cancer vaccines aim to target personalized neoantigens, inducing strong and specific anti-tumour immune responses, and have garnered widespread attention for their tremendous potential in cancer prevention and treatment. Compared to traditional cancer vaccine platforms, mRNA cancer vaccines offer unique advantages such as high safety, strong immunogenicity, personalized targeted immunotherapy, shorter development cycles, and lower costs, making them a revolutionary tool for achieving the goals of precision medicine. Given the unique advantages of MM, such as antigen lineage specificity, pathological mechanism and therapeutic needs alignment, clear safety profile and treatment endpoints, clinical window, and compatibility with multi-strategy combination therapies, we believe that exploring the use of mRNA cancer vaccines for the treatment of MM is both necessary and promising. In this review, we aimed to summarize the progress of immunotherapy for MM and the current application status of various antigens and vaccines. On this basis, we analysed the specificity and feasibility of mRNA cancer vaccines in MM and proposed strategies for their clinical management and optimisation. We believe that mRNA cancer vaccines have the potential to redefine the treatment paradigm for MM, offering new solutions for patients with refractory and relapsed disease.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1880 6","pages":"Article 189493"},"PeriodicalIF":9.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145491067","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The double-edged sword of IRF7: “Guardian” or “traitor” of tumors? From mechanistic contradictions to therapeutic revolution IRF7的双刃剑:肿瘤的“守护者”还是“叛徒”?从机械矛盾到治疗革命。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-01 DOI: 10.1016/j.bbcan.2025.189501
Siyu Li , Qiong Luo , Jiali Hu , Xiaobing Duan , Guihai Zhang

Abstract

Interferon Regulatory Factor 7 (IRF7) is emerging as a pivotal and paradoxical modulator in cancer biology, functioning both as a tumor suppressor and as a context-dependent oncogenic driver. This review not only covers classical features such as proliferative signaling, immune evasion, apoptosis resistance, and metastasis but also includes a dedicated discussion of the four emerging features added by Hanahan in 2022, namely, phenotypic plasticity, nonmutational epigenetic reprogramming, polymorphic microbiomes, and the dynamic balance of senescent cells. Mechanistically, IRF7 integrates transcriptional regulation, Posttranslational Modifications (PTMs), and microenvironmental cues to orchestrate diverse oncogenic processes, resulting in tissue-specific and spatiotemporal functional dichotomy. Furthermore, this review highlights the dualistic potential of IRF7 in therapeutic contexts—enhancing antitumor immunity and interferon signaling in some settings while contributing to immune suppression, therapeutic resistance, or tumor progression in others. Recent advances in drug development targeting IRF7, such as epigenetic modulation, kinase inhibition, and exosome-based delivery, suggest promising avenues for precision cancer therapy. However, the functional heterogeneity of IRF7 necessitates a nuanced therapeutic approach, emphasizing biomarker-guided stratification and spatiotemporal-specific interventions. Overall, IRF7 represents a key regulatory hub linked to immune, metabolic, and oncogenic pathways, with significant implications for future cancer diagnosis, prognosis, and treatment strategies.
干扰素调节因子7 (IRF7)正在成为癌症生物学中一个关键而矛盾的调节剂,既可以作为肿瘤抑制因子,也可以作为环境依赖性的致癌驱动因子。这篇综述不仅涵盖了增殖信号、免疫逃避、细胞凋亡抵抗和转移等经典特征,而且还包括了Hanahan在2022年增加的四个新兴特征,即表型可塑性、非突变表观遗传重编程、多态微生物组和衰老细胞的动态平衡。在机制上,IRF7整合了转录调控、翻译后修饰(PTMs)和微环境线索来协调不同的致癌过程,导致组织特异性和时空功能二分法。此外,本综述强调了IRF7在治疗环境中的双重潜力——在某些情况下增强抗肿瘤免疫和干扰素信号,而在其他情况下促进免疫抑制、治疗抵抗或肿瘤进展。最近针对IRF7的药物开发进展,如表观遗传调节、激酶抑制和基于外泌体的递送,为精确癌症治疗提供了有希望的途径。然而,IRF7的功能异质性需要一种细致入微的治疗方法,强调生物标志物引导的分层和时空特异性干预。总的来说,IRF7是一个与免疫、代谢和致癌途径相关的关键调控枢纽,对未来的癌症诊断、预后和治疗策略具有重要意义。
{"title":"The double-edged sword of IRF7: “Guardian” or “traitor” of tumors? From mechanistic contradictions to therapeutic revolution","authors":"Siyu Li ,&nbsp;Qiong Luo ,&nbsp;Jiali Hu ,&nbsp;Xiaobing Duan ,&nbsp;Guihai Zhang","doi":"10.1016/j.bbcan.2025.189501","DOIUrl":"10.1016/j.bbcan.2025.189501","url":null,"abstract":"<div><h3>Abstract</h3><div>Interferon Regulatory Factor 7 (IRF7) is emerging as a pivotal and paradoxical modulator in cancer biology, functioning both as a tumor suppressor and as a context-dependent oncogenic driver. This review not only covers classical features such as proliferative signaling, immune evasion, apoptosis resistance, and metastasis but also includes a dedicated discussion of the four emerging features added by Hanahan in 2022, namely, phenotypic plasticity, nonmutational epigenetic reprogramming, polymorphic microbiomes, and the dynamic balance of senescent cells. Mechanistically, IRF7 integrates transcriptional regulation, Posttranslational Modifications (PTMs), and microenvironmental cues to orchestrate diverse oncogenic processes, resulting in tissue-specific and spatiotemporal functional dichotomy. Furthermore, this review highlights the dualistic potential of IRF7 in therapeutic contexts—enhancing antitumor immunity and interferon signaling in some settings while contributing to immune suppression, therapeutic resistance, or tumor progression in others. Recent advances in drug development targeting IRF7, such as epigenetic modulation, kinase inhibition, and exosome-based delivery, suggest promising avenues for precision cancer therapy. However, the functional heterogeneity of IRF7 necessitates a nuanced therapeutic approach, emphasizing biomarker-guided stratification and spatiotemporal-specific interventions. Overall, IRF7 represents a key regulatory hub linked to immune, metabolic, and oncogenic pathways, with significant implications for future cancer diagnosis, prognosis, and treatment strategies.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1880 6","pages":"Article 189501"},"PeriodicalIF":9.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145596993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Role of CCL8 in cancer immunity and tumor progression: Implications for cancer treatment CCL8在癌症免疫和肿瘤进展中的作用:对癌症治疗的意义
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-11-01 DOI: 10.1016/j.bbcan.2025.189502
Jiwoo Jeong, Eun-Sook Kim
The C-C motif chemokine ligand 8 (CCL8) is a key chemokine that plays a prominent role in immune cells, inflammatory responses, and the progression of various pathologies. This review initially outlines the structural characteristics of CCL8, its receptor-binding properties, and its involvement in various signaling pathways. We integrated database evidence demonstrating how CCL8 expression in various cancers affects tumor growth, migration, and invasion, thereby influencing patient prognosis. Subsequently, we explored how CCL8 interacts with immune cells to either promote or suppress tumor development and assessed whether its expression correlates with immune cell infiltration within the tumor immune microenvironment in different cancer types. Furthermore, we underscore the potential of CCL8 as a biomarker for the progression of cancer and other diseases, and its clinical relevance in predicting therapeutic responses via targeted antagonists and receptor blockade. Finally, by integrating current insights into the role of CCL8 in tumor cell proliferation within the cancer immune microenvironment, this review aims to elucidate the biological significance of CCL8 and facilitate the development of novel diagnostic and therapeutic strategies.
C-C基序趋化因子配体8 (CCL8)是一种关键的趋化因子,在免疫细胞、炎症反应和各种病理进展中发挥着重要作用。本文首先概述了CCL8的结构特征、受体结合特性及其参与的各种信号通路。我们整合了数据库证据,证明CCL8在各种癌症中的表达如何影响肿瘤生长、迁移和侵袭,从而影响患者预后。随后,我们探索了CCL8如何与免疫细胞相互作用,促进或抑制肿瘤的发展,并评估了其表达是否与不同癌症类型肿瘤免疫微环境中的免疫细胞浸润相关。此外,我们强调了CCL8作为癌症和其他疾病进展的生物标志物的潜力,以及它在通过靶向拮抗剂和受体阻断预测治疗反应方面的临床相关性。最后,通过整合目前对肿瘤免疫微环境中CCL8在肿瘤细胞增殖中的作用的见解,本综述旨在阐明CCL8的生物学意义,并促进新的诊断和治疗策略的发展。
{"title":"Role of CCL8 in cancer immunity and tumor progression: Implications for cancer treatment","authors":"Jiwoo Jeong,&nbsp;Eun-Sook Kim","doi":"10.1016/j.bbcan.2025.189502","DOIUrl":"10.1016/j.bbcan.2025.189502","url":null,"abstract":"<div><div>The C-C motif chemokine ligand 8 (CCL8) is a key chemokine that plays a prominent role in immune cells, inflammatory responses, and the progression of various pathologies. This review initially outlines the structural characteristics of CCL8, its receptor-binding properties, and its involvement in various signaling pathways. We integrated database evidence demonstrating how CCL8 expression in various cancers affects tumor growth, migration, and invasion, thereby influencing patient prognosis. Subsequently, we explored how CCL8 interacts with immune cells to either promote or suppress tumor development and assessed whether its expression correlates with immune cell infiltration within the tumor immune microenvironment in different cancer types. Furthermore, we underscore the potential of CCL8 as a biomarker for the progression of cancer and other diseases, and its clinical relevance in predicting therapeutic responses via targeted antagonists and receptor blockade. Finally, by integrating current insights into the role of CCL8 in tumor cell proliferation within the cancer immune microenvironment, this review aims to elucidate the biological significance of CCL8 and facilitate the development of novel diagnostic and therapeutic strategies.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1880 6","pages":"Article 189502"},"PeriodicalIF":9.7,"publicationDate":"2025-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145643916","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Revisiting CHK1 and WEE1 kinases as therapeutic targets in cancer 重新审视CHK1和WEE1激酶作为癌症治疗靶点的作用
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-22 DOI: 10.1016/j.bbcan.2025.189482
Nehal Arvind Kumar, P. Manasa, Jaikanth Chandrasekaran
Uncontrolled cell division is a defining characteristic of cancer, driven by genomic instability. This instability disrupts the DNA damage response network, exacerbates replication stress, and impairs cell cycle checkpoints. Although conventional DNA-damaging therapies like chemotherapy and radiotherapy exploit this vulnerability, they often cause substantial damage to healthy tissues, leading to adverse effects that may inadvertently promote tumour resilience. Targeted DDR inhibition, exemplified by Poly [ADP-ribose] polymerase inhibitors, has achieved major clinical success, functioning through a synthetic lethality approach in tumours with a defective homologous recombination repair pathway. Extending this strategy to the ATR-CHK1-WEE1 axis, a central responder to replication stress and regulator of intra-S and G2/M checkpoints, offers a promising therapeutic window, particularly in cancers with G1/S checkpoint defects. However, realising this potential requires a deeper understanding of tumour-specific DNA damage response defects, identification of robust biomarkers, and improved patient stratification needs. Currently, the clinical development of CHK1 and WEE1 inhibitors demonstrates their potential, especially in combination with standard-of-care therapies, where they capitalise on elevated replication stress to induce selective tumour cell death. This review outlines recent advances, challenges, and future directions in targeting CHK1 and WEE1 kinases, highlighting their potential to advance precision oncology in the future.
不受控制的细胞分裂是癌症的一个决定性特征,由基因组不稳定驱动。这种不稳定性破坏了DNA损伤反应网络,加剧了复制压力,并损害了细胞周期检查点。尽管化疗和放疗等传统的dna损伤疗法利用了这种脆弱性,但它们往往会对健康组织造成实质性损害,导致可能无意中促进肿瘤恢复能力的不利影响。以聚[adp -核糖]聚合酶抑制剂为例,靶向DDR抑制已经取得了重大的临床成功,通过合成致死性方法在具有缺陷同源重组修复途径的肿瘤中发挥作用。将该策略扩展到ATR-CHK1-WEE1轴,这是复制应激的中心应答者和S内和G2/M检查点的调节剂,提供了一个有希望的治疗窗口,特别是在G1/S检查点缺陷的癌症中。然而,实现这一潜力需要更深入地了解肿瘤特异性DNA损伤反应缺陷,识别强大的生物标志物,并改善患者分层需求。目前,CHK1和WEE1抑制剂的临床发展证明了它们的潜力,特别是在与标准治疗相结合的情况下,它们利用升高的复制应激来诱导选择性肿瘤细胞死亡。本文概述了靶向CHK1和WEE1激酶的最新进展、挑战和未来方向,强调了它们在未来推进精确肿瘤学的潜力。
{"title":"Revisiting CHK1 and WEE1 kinases as therapeutic targets in cancer","authors":"Nehal Arvind Kumar,&nbsp;P. Manasa,&nbsp;Jaikanth Chandrasekaran","doi":"10.1016/j.bbcan.2025.189482","DOIUrl":"10.1016/j.bbcan.2025.189482","url":null,"abstract":"<div><div>Uncontrolled cell division is a defining characteristic of cancer, driven by genomic instability. This instability disrupts the DNA damage response network, exacerbates replication stress, and impairs cell cycle checkpoints. Although conventional DNA-damaging therapies like chemotherapy and radiotherapy exploit this vulnerability, they often cause substantial damage to healthy tissues, leading to adverse effects that may inadvertently promote tumour resilience. Targeted DDR inhibition, exemplified by Poly [ADP-ribose] polymerase inhibitors, has achieved major clinical success, functioning through a synthetic lethality approach in tumours with a defective homologous recombination repair pathway. Extending this strategy to the ATR-CHK1-WEE1 axis, a central responder to replication stress and regulator of intra-S and G2/M checkpoints, offers a promising therapeutic window, particularly in cancers with G1/S checkpoint defects. However, realising this potential requires a deeper understanding of tumour-specific DNA damage response defects, identification of robust biomarkers, and improved patient stratification needs. Currently, the clinical development of CHK1 and WEE1 inhibitors demonstrates their potential, especially in combination with standard-of-care therapies, where they capitalise on elevated replication stress to induce selective tumour cell death. This review outlines recent advances, challenges, and future directions in targeting CHK1 and WEE1 kinases, highlighting their potential to advance precision oncology in the future.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1880 6","pages":"Article 189482"},"PeriodicalIF":9.7,"publicationDate":"2025-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145358994","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Research progress on gut microbiota in colorectal cancer immunotherapy 肠道菌群在结直肠癌免疫治疗中的研究进展。
IF 9.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-10-21 DOI: 10.1016/j.bbcan.2025.189476
Jing Li , Yingkun Yue , Jiaxin Pan , Fang Liang
Immune checkpoint inhibitors (ICIs) have demonstrated significant clinical benefits in treating various malignancies. However, their therapeutic efficacy exhibits considerable interindividual variability in patients with colorectal cancer (CRC). In recent years, growing attention has been focused on the regulatory role of the gut microbiota and its metabolic microenvironment in modulating ICIs responses. This article systematically reviews key advances in understanding how the gut microbiota and its metabolites influence ICIs efficacy. For example: Specific bacterial species (e.g., Lactobacillus paracasei and Fusobacterium nucleatum) may regulate ICIs efficacy by modulating antigen presentation or the tumor immune microenvironment. Microbial metabolites, such as short-chain fatty acids (SCFAs), can enhance immune function and thereby improve ICIs outcomes. Potential microbiome-targeted interventions—including probiotic/prebiotic combinations, optimized antibiotic administration timing, refined fecal microbiota transplantation (FMT) protocols, and engineered synthetic biology-based bacterial therapies—are also discussed. By synthesizing current evidence, this review provides a theoretical foundation for developing novel personalized immunotherapy strategies for CRC, with a focus on microbiome modulation to optimize ICIs treatment efficacy.
免疫检查点抑制剂(ICIs)在治疗各种恶性肿瘤方面已显示出显著的临床疗效。然而,它们的治疗效果在结直肠癌(CRC)患者中表现出相当大的个体差异。近年来,人们越来越关注肠道微生物群及其代谢微环境在调节ICIs反应中的调节作用。本文系统地回顾了了解肠道微生物群及其代谢物如何影响ICIs疗效的关键进展。例如:特定的细菌种类(如副干酪乳杆菌和核梭杆菌)可能通过调节抗原呈递或肿瘤免疫微环境来调节ICIs的疗效。微生物代谢物,如短链脂肪酸(SCFAs),可以增强免疫功能,从而改善ICIs的预后。潜在的以微生物群为目标的干预措施,包括益生菌/益生元组合、优化抗生素给药时间、改进的粪便微生物群移植(FMT)方案和基于工程合成生物学的细菌治疗也进行了讨论。通过综合现有证据,本综述为开发新的CRC个性化免疫治疗策略提供了理论基础,重点关注微生物组调节以优化ICIs的治疗效果。
{"title":"Research progress on gut microbiota in colorectal cancer immunotherapy","authors":"Jing Li ,&nbsp;Yingkun Yue ,&nbsp;Jiaxin Pan ,&nbsp;Fang Liang","doi":"10.1016/j.bbcan.2025.189476","DOIUrl":"10.1016/j.bbcan.2025.189476","url":null,"abstract":"<div><div>Immune checkpoint inhibitors (ICIs) have demonstrated significant clinical benefits in treating various malignancies. However, their therapeutic efficacy exhibits considerable interindividual variability in patients with colorectal cancer (CRC). In recent years, growing attention has been focused on the regulatory role of the gut microbiota and its metabolic microenvironment in modulating ICIs responses. This article systematically reviews key advances in understanding how the gut microbiota and its metabolites influence ICIs efficacy. For example: Specific bacterial species (e.g., <em>Lactobacillus paracasei</em> and <em>Fusobacterium nucleatum</em>) may regulate ICIs efficacy by modulating antigen presentation or the tumor immune microenvironment. Microbial metabolites, such as short-chain fatty acids (SCFAs), can enhance immune function and thereby improve ICIs outcomes. Potential microbiome-targeted interventions—including probiotic/prebiotic combinations, optimized antibiotic administration timing, refined fecal microbiota transplantation (FMT) protocols, and engineered synthetic biology-based bacterial therapies—are also discussed. By synthesizing current evidence, this review provides a theoretical foundation for developing novel personalized immunotherapy strategies for CRC, with a focus on microbiome modulation to optimize ICIs treatment efficacy.</div></div>","PeriodicalId":8782,"journal":{"name":"Biochimica et biophysica acta. Reviews on cancer","volume":"1880 6","pages":"Article 189476"},"PeriodicalIF":9.7,"publicationDate":"2025-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145357156","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Biochimica et biophysica acta. Reviews on cancer
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号: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