首页 > 最新文献

International review of cell and molecular biology最新文献

英文 中文
Genetic and epigenetic features of neuroendocrine prostate cancer and their emerging applications. 神经内分泌前列腺癌的遗传和表观遗传特征及其新兴应用。
3区 生物学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-01 Epub Date: 2023-07-07 DOI: 10.1016/bs.ircmb.2023.06.002
Xintong Zhang, Edward Barnett, Jim Smith, Emma Wilkinson, Rathan M Subramaniam, Amir Zarrabi, Euan J Rodger, Aniruddha Chatterjee

Prostate cancer is the second most prevalent cancer in men globally. De novo neuroendocrine prostate cancer (NEPC) is uncommon at initial diagnosis, however, (treatment-induced) t-NEPC emerges in up to 25% of prostate adenocarcinoma (PRAD) cases treated with androgen deprivation, carrying a drastically poor prognosis. The transition from PRAD to t-NEPC is underpinned by several key genetic mutations; TP53, RB1, and MYCN are the main genes implicated, bearing similarities to other neuroendocrine tumours. A broad range of epigenetic alterations, such as aberrations in DNA methylation, histone post-translational modifications, and non-coding RNAs, may drive lineage plasticity from PRAD to t-NEPC. The clinical diagnosis of NEPC is hampered by a lack of accessible biomarkers; recent advances in liquid biopsy techniques assessing circulating tumour cells and ctDNA in NEPC suggest that the advent of non-invasive means of monitoring progression to NEPC is on the horizon. Such techniques are vital for NEPC management; diagnosis of t-NEPC is crucial for implementing effective treatment, and precision medicine will be integral to providing the best outcomes for patients.

前列腺癌是全球男性发病率第二高的癌症。新发神经内分泌性前列腺癌(NEPC)在最初诊断时并不常见,但在接受雄激素剥夺治疗的前列腺腺癌(PRAD)病例中,高达 25% 的病例会出现(治疗诱发的)t-NEPC,预后极差。从 PRAD 到 t-NEPC 的转变由几个关键基因突变支撑;TP53、RB1 和 MYCN 是主要的相关基因,与其他神经内分泌肿瘤有相似之处。广泛的表观遗传学改变,如DNA甲基化、组蛋白翻译后修饰和非编码RNA的畸变,可能会驱动从PRAD到t-NEPC的系谱可塑性。NEPC 的临床诊断因缺乏可获得的生物标志物而受到阻碍;评估 NEPC 循环肿瘤细胞和 ctDNA 的液体活检技术的最新进展表明,监测 NEPC 进展的无创手段即将出现。这些技术对 NEPC 的管理至关重要;t-NEPC 的诊断对实施有效治疗至关重要,而精准医疗将是为患者提供最佳治疗效果不可或缺的一部分。
{"title":"Genetic and epigenetic features of neuroendocrine prostate cancer and their emerging applications.","authors":"Xintong Zhang, Edward Barnett, Jim Smith, Emma Wilkinson, Rathan M Subramaniam, Amir Zarrabi, Euan J Rodger, Aniruddha Chatterjee","doi":"10.1016/bs.ircmb.2023.06.002","DOIUrl":"10.1016/bs.ircmb.2023.06.002","url":null,"abstract":"<p><p>Prostate cancer is the second most prevalent cancer in men globally. De novo neuroendocrine prostate cancer (NEPC) is uncommon at initial diagnosis, however, (treatment-induced) t-NEPC emerges in up to 25% of prostate adenocarcinoma (PRAD) cases treated with androgen deprivation, carrying a drastically poor prognosis. The transition from PRAD to t-NEPC is underpinned by several key genetic mutations; TP53, RB1, and MYCN are the main genes implicated, bearing similarities to other neuroendocrine tumours. A broad range of epigenetic alterations, such as aberrations in DNA methylation, histone post-translational modifications, and non-coding RNAs, may drive lineage plasticity from PRAD to t-NEPC. The clinical diagnosis of NEPC is hampered by a lack of accessible biomarkers; recent advances in liquid biopsy techniques assessing circulating tumour cells and ctDNA in NEPC suggest that the advent of non-invasive means of monitoring progression to NEPC is on the horizon. Such techniques are vital for NEPC management; diagnosis of t-NEPC is crucial for implementing effective treatment, and precision medicine will be integral to providing the best outcomes for patients.</p>","PeriodicalId":14422,"journal":{"name":"International review of cell and molecular biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139740925","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epigenetic control of cell signalling in cancer stem cells. 癌症干细胞中细胞信号的表观遗传控制。
3区 生物学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-01 Epub Date: 2023-08-17 DOI: 10.1016/bs.ircmb.2023.07.009
Gaurav Joshi, Amitava Basu

The self-renewing cancer stem cells (CSCs) represent one of the distinct cell populations occurring in a tumour that can differentiate into multiple lineages. This group of sparsely abundant cells play a vital role in tumour survival and resistance to different treatments during cancer. The lack of exclusive markers associated with CSCs makes diagnosis and prognosis in cancer patients extremely difficult. This calls for the identification of unique regulators and markers for CSCs. Various signalling pathways like the Wnt/β-catenin pathway, Hedgehog pathway, Notch pathway, and TGFβ/BMP play a major role in the regulation and maintenance of CSCs. Epigenetic regulatory mechanisms add another layer of complexity to control these signalling pathways. In this chapter, we discuss about the role of epigenetic mechanisms in regulating the cellular signalling pathways in CSCs. The epigenetic regulatory mechanisms such as DNA methylation, histone modification and microRNAs can modulate the diverse effectors of signalling pathways and consequently the growth, differentiation and tumorigenicity of CSCs. In the end, we briefly discuss the therapeutic potential of targeting these epigenetic regulators and their target genes in CSCs.

自我更新的癌症干细胞(CSCs)是肿瘤中出现的独特细胞群之一,可分化成多种细胞系。这群数量稀少的细胞在肿瘤存活和抗癌过程中对不同治疗起着至关重要的作用。由于缺乏与 CSC 相关的专属标记物,癌症患者的诊断和预后极为困难。这就需要确定 CSCs 的独特调节因子和标记物。Wnt/β-catenin通路、刺猬通路、Notch通路和TGFβ/BMP等各种信号通路在调控和维持CSCs方面发挥着重要作用。表观遗传调控机制为控制这些信号通路增加了另一层复杂性。在本章中,我们将讨论表观遗传机制在调控 CSCs 细胞信号通路中的作用。DNA甲基化、组蛋白修饰和microRNA等表观遗传调控机制可调节信号通路的多种效应因子,进而影响CSCs的生长、分化和致瘤性。最后,我们简要讨论了针对这些表观遗传调控因子及其靶基因在 CSCs 中的治疗潜力。
{"title":"Epigenetic control of cell signalling in cancer stem cells.","authors":"Gaurav Joshi, Amitava Basu","doi":"10.1016/bs.ircmb.2023.07.009","DOIUrl":"10.1016/bs.ircmb.2023.07.009","url":null,"abstract":"<p><p>The self-renewing cancer stem cells (CSCs) represent one of the distinct cell populations occurring in a tumour that can differentiate into multiple lineages. This group of sparsely abundant cells play a vital role in tumour survival and resistance to different treatments during cancer. The lack of exclusive markers associated with CSCs makes diagnosis and prognosis in cancer patients extremely difficult. This calls for the identification of unique regulators and markers for CSCs. Various signalling pathways like the Wnt/β-catenin pathway, Hedgehog pathway, Notch pathway, and TGFβ/BMP play a major role in the regulation and maintenance of CSCs. Epigenetic regulatory mechanisms add another layer of complexity to control these signalling pathways. In this chapter, we discuss about the role of epigenetic mechanisms in regulating the cellular signalling pathways in CSCs. The epigenetic regulatory mechanisms such as DNA methylation, histone modification and microRNAs can modulate the diverse effectors of signalling pathways and consequently the growth, differentiation and tumorigenicity of CSCs. In the end, we briefly discuss the therapeutic potential of targeting these epigenetic regulators and their target genes in CSCs.</p>","PeriodicalId":14422,"journal":{"name":"International review of cell and molecular biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139740996","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epigenetic therapeutic strategies in pancreatic cancer. 胰腺癌的表观遗传学治疗策略。
3区 生物学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-01 Epub Date: 2024-01-24 DOI: 10.1016/bs.ircmb.2023.10.002
Arturo Orlacchio, Stephen Muzyka, Tamas A Gonda

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid malignancies, characterized by its aggressiveness and metastatic potential, with a 5-year survival rate of only 8-11%. Despite significant improvements in PDAC treatment and management, therapeutic alternatives are still limited. One of the main reasons is its high degree of intra- and inter-individual tumor heterogeneity which is established and maintained through a complex network of transcription factors and epigenetic regulators. Epigenetic drugs, have shown promising preclinical results in PDAC and are currently being evaluated in clinical trials both for their ability to sensitize cancer cells to cytotoxic drugs and to counteract the immunosuppressive characteristic of PDAC tumor microenvironment. In this review, we discuss the current status of epigenetic treatment strategies to overcome molecular and cellular PDAC heterogeneity in order to improve response to therapy.

胰腺导管腺癌(PDAC)是最致命的实体恶性肿瘤之一,具有侵袭性和转移性,5 年生存率仅为 8-11%。尽管在 PDAC 的治疗和管理方面取得了重大进展,但替代疗法仍然有限。其中一个主要原因是其个体内和个体间肿瘤的高度异质性,这种异质性是通过转录因子和表观遗传调节因子的复杂网络建立和维持的。表观遗传药物在 PDAC 的临床前研究中取得了可喜的成果,目前正在临床试验中对其进行评估,以确定其是否能使癌细胞对细胞毒性药物敏感,以及是否能抵消 PDAC 肿瘤微环境的免疫抑制特性。在本综述中,我们将讨论表观遗传学治疗策略的现状,以克服 PDAC 的分子和细胞异质性,从而改善治疗反应。
{"title":"Epigenetic therapeutic strategies in pancreatic cancer.","authors":"Arturo Orlacchio, Stephen Muzyka, Tamas A Gonda","doi":"10.1016/bs.ircmb.2023.10.002","DOIUrl":"10.1016/bs.ircmb.2023.10.002","url":null,"abstract":"<p><p>Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid malignancies, characterized by its aggressiveness and metastatic potential, with a 5-year survival rate of only 8-11%. Despite significant improvements in PDAC treatment and management, therapeutic alternatives are still limited. One of the main reasons is its high degree of intra- and inter-individual tumor heterogeneity which is established and maintained through a complex network of transcription factors and epigenetic regulators. Epigenetic drugs, have shown promising preclinical results in PDAC and are currently being evaluated in clinical trials both for their ability to sensitize cancer cells to cytotoxic drugs and to counteract the immunosuppressive characteristic of PDAC tumor microenvironment. In this review, we discuss the current status of epigenetic treatment strategies to overcome molecular and cellular PDAC heterogeneity in order to improve response to therapy.</p>","PeriodicalId":14422,"journal":{"name":"International review of cell and molecular biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139740998","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Preconditioning with immunogenic cell death-inducing treatments for subsequent immunotherapy. 用诱导免疫细胞死亡的预处理方法进行后续免疫疗法。
3区 生物学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-01 Epub Date: 2023-07-26 DOI: 10.1016/bs.ircmb.2023.06.001
Hui Pan, Peng Liu, Guido Kroemer, Oliver Kepp

Since the dawn of anticancer immunotherapy, the clinical use of immune checkpoint inhibitors (ICI) has increased exponentially. Monoclonal antibodies targeting CTLA-4 and the PD-1/PD-L1 interaction were first introduced for the treatment of patients with unresectable melanoma. In melanoma, ICI lead to durable regression in a significant number of patients and have thus been clinically approved as a first-line treatment of advanced disease. Over the past years an increasing number of regulatory approvals have been granted for the use of ICI in patients affected by a large range of distinct carcinomas. In retrospect surprisingly, it has been discovered that particularly successful chemotherapeutic treatments are able to trigger anticancer immune responses because they induce immunogenic cell death (ICD), hence killing cancer cells in a way that they elicit an immune response against tumor-associated antigens. Logically, preclinical studies as well as clinical trials are currently exploring the possibility to combine ICD inducers with ICI to obtain optimal therapeutic effects. Here, we provide a broad overview of current strategies for the implementation of combinatorial approaches involving ICD induction followed by ICI in anticancer therapy.

自抗癌免疫疗法诞生以来,免疫检查点抑制剂(ICI)的临床应用呈指数级增长。针对 CTLA-4 和 PD-1/PD-L1 相互作用的单克隆抗体首次被用于治疗无法切除的黑色素瘤患者。在黑色素瘤治疗中,ICI 可使大量患者的病情得到持久缓解,因此被临床批准作为晚期疾病的一线治疗药物。在过去几年中,越来越多的监管机构批准将 ICI 用于多种不同的癌症患者。回过头来看,人们惊讶地发现,特别成功的化疗方法能够引发抗癌免疫反应,因为它们能诱导免疫原性细胞死亡(ICD),从而杀死癌细胞,引起针对肿瘤相关抗原的免疫反应。从逻辑上讲,临床前研究和临床试验目前都在探索将 ICD 诱导剂与 ICI 结合使用以获得最佳治疗效果的可能性。在此,我们将概述目前在抗癌治疗中实施 ICD 诱导和 ICI 组合疗法的策略。
{"title":"Preconditioning with immunogenic cell death-inducing treatments for subsequent immunotherapy.","authors":"Hui Pan, Peng Liu, Guido Kroemer, Oliver Kepp","doi":"10.1016/bs.ircmb.2023.06.001","DOIUrl":"10.1016/bs.ircmb.2023.06.001","url":null,"abstract":"<p><p>Since the dawn of anticancer immunotherapy, the clinical use of immune checkpoint inhibitors (ICI) has increased exponentially. Monoclonal antibodies targeting CTLA-4 and the PD-1/PD-L1 interaction were first introduced for the treatment of patients with unresectable melanoma. In melanoma, ICI lead to durable regression in a significant number of patients and have thus been clinically approved as a first-line treatment of advanced disease. Over the past years an increasing number of regulatory approvals have been granted for the use of ICI in patients affected by a large range of distinct carcinomas. In retrospect surprisingly, it has been discovered that particularly successful chemotherapeutic treatments are able to trigger anticancer immune responses because they induce immunogenic cell death (ICD), hence killing cancer cells in a way that they elicit an immune response against tumor-associated antigens. Logically, preclinical studies as well as clinical trials are currently exploring the possibility to combine ICD inducers with ICI to obtain optimal therapeutic effects. Here, we provide a broad overview of current strategies for the implementation of combinatorial approaches involving ICD induction followed by ICI in anticancer therapy.</p>","PeriodicalId":14422,"journal":{"name":"International review of cell and molecular biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139472122","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Immunological landscape of solid cancer: Interplay between tumor and autoimmunity. 实体瘤的免疫格局:肿瘤与自身免疫之间的相互作用
3区 生物学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-01 Epub Date: 2024-05-31 DOI: 10.1016/bs.ircmb.2024.04.002
Ajay K Singh, Phaneendra K Duddempudi, Divya Borasandra Kenchappa, Nityanand Srivastava, Nitin P Amdare

The immune system, a central player in maintaining homeostasis, emerges as a pivotal factor in the pathogenesis and progression of two seemingly disparate yet interconnected categories of diseases: autoimmunity and cancer. This chapter delves into the intricate and multifaceted role of the immune system, particularly T cells, in orchestrating responses that govern the delicate balance between immune surveillance and self-tolerance. T cells, pivotal immune system components, play a central role in both diseases. In autoimmunity, aberrant T cell activation drives damaging immune responses against normal tissues, while in cancer, T cells exhibit suppressed responses, allowing the growth of malignant tumors. Immune checkpoint receptors, example, initially explored in autoimmunity, now revolutionize cancer treatment via immune checkpoint blockade (ICB). Though effective in various tumors, ICB poses risks of immune-related adverse events (irAEs) akin to autoimmunity. This chapter underscores the importance of understanding tumor-associated antigens and their role in autoimmunity, immune checkpoint regulation, and their implications for both diseases. It also explores autoimmunity resulting from cancer immunotherapy and shared molecular pathways in solid tumors and autoimmune diseases, highlighting their interconnectedness at the molecular level. Additionally, it sheds light on common pathways and epigenetic features shared by autoimmunity and cancer, and the potential of repurposing drugs for therapeutic interventions. Delving deeper into these insights could unlock therapeutic strategies for both autoimmunity and cancer.

免疫系统是维持体内平衡的核心角色,在自身免疫和癌症这两类看似不同却又相互关联的疾病的发病和发展过程中,免疫系统扮演着举足轻重的角色。本章将深入探讨免疫系统,尤其是 T 细胞,在协调免疫监视和自我耐受之间微妙平衡的反应中发挥的错综复杂的多方面作用。T 细胞是免疫系统的关键组成部分,在这两种疾病中都发挥着核心作用。在自身免疫病中,异常的 T 细胞活化驱动着针对正常组织的破坏性免疫反应;而在癌症中,T 细胞则表现出抑制性反应,使恶性肿瘤得以生长。例如,免疫检查点受体最初是在自身免疫病中探索出来的,现在则通过免疫检查点阻断(ICB)彻底改变了癌症治疗。虽然 ICB 对各种肿瘤有效,但它也带来了与自身免疫类似的免疫相关不良事件(irAEs)的风险。本章强调了了解肿瘤相关抗原及其在自身免疫中的作用、免疫检查点调控及其对这两种疾病的影响的重要性。本章还探讨了癌症免疫疗法导致的自身免疫以及实体瘤和自身免疫性疾病的共同分子通路,强调了它们在分子水平上的相互联系。此外,它还揭示了自身免疫和癌症的共同途径和表观遗传学特征,以及重新利用药物进行治疗干预的潜力。深入研究这些见解可以为自身免疫病和癌症找到治疗策略。
{"title":"Immunological landscape of solid cancer: Interplay between tumor and autoimmunity.","authors":"Ajay K Singh, Phaneendra K Duddempudi, Divya Borasandra Kenchappa, Nityanand Srivastava, Nitin P Amdare","doi":"10.1016/bs.ircmb.2024.04.002","DOIUrl":"https://doi.org/10.1016/bs.ircmb.2024.04.002","url":null,"abstract":"<p><p>The immune system, a central player in maintaining homeostasis, emerges as a pivotal factor in the pathogenesis and progression of two seemingly disparate yet interconnected categories of diseases: autoimmunity and cancer. This chapter delves into the intricate and multifaceted role of the immune system, particularly T cells, in orchestrating responses that govern the delicate balance between immune surveillance and self-tolerance. T cells, pivotal immune system components, play a central role in both diseases. In autoimmunity, aberrant T cell activation drives damaging immune responses against normal tissues, while in cancer, T cells exhibit suppressed responses, allowing the growth of malignant tumors. Immune checkpoint receptors, example, initially explored in autoimmunity, now revolutionize cancer treatment via immune checkpoint blockade (ICB). Though effective in various tumors, ICB poses risks of immune-related adverse events (irAEs) akin to autoimmunity. This chapter underscores the importance of understanding tumor-associated antigens and their role in autoimmunity, immune checkpoint regulation, and their implications for both diseases. It also explores autoimmunity resulting from cancer immunotherapy and shared molecular pathways in solid tumors and autoimmune diseases, highlighting their interconnectedness at the molecular level. Additionally, it sheds light on common pathways and epigenetic features shared by autoimmunity and cancer, and the potential of repurposing drugs for therapeutic interventions. Delving deeper into these insights could unlock therapeutic strategies for both autoimmunity and cancer.</p>","PeriodicalId":14422,"journal":{"name":"International review of cell and molecular biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142465575","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
DNA damage response and neoantigens: A favorable target for triple-negative breast cancer immunotherapy and vaccine development. DNA 损伤反应和新抗原:三阴性乳腺癌免疫疗法和疫苗开发的有利靶点。
3区 生物学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-01 Epub Date: 2024-06-04 DOI: 10.1016/bs.ircmb.2024.05.001
Rajasekaran Subbarayan, Dhasarathdev Srinivasan, Ranjith Balakrishnan, Ajeet Kumar, Salman Sadullah Usmani, Nityanand Srivastava

Triple-negative breast cancer (TNBC) poses a significant clinical challenge due to its aggressive nature and limited therapeutic options. The interplay between DNA damage response (DDR) mechanisms and the emergence of neoantigens represents a promising avenue for developing targeted immunotherapeutic strategies and vaccines for TNBC. The DDR is a complex network of cellular mechanisms designed to maintain genomic integrity. In TNBC, where genetic instability is a hallmark, dysregulation of DDR components plays a pivotal role in tumorigenesis and progression. This review explores the intricate relationship between DDR and neoantigens, shedding light on the potential vulnerabilities of TNBC cells. Neoantigens, arising from somatic mutations in cancer cells, represent unique antigens that can be recognized by the immune system. TNBC's propensity for genomic instability leads to an increased mutational burden, consequently yielding a rich repertoire of neoantigens. The convergence of DDR and neoantigens in TNBC offers a distinctive opportunity for immunotherapeutic targeting. Immunotherapy has revolutionized cancer treatment by harnessing the immune system to selectively target cancer cells. The unique immunogenicity conferred by DDR-related neoantigens in TNBC positions them as ideal targets for immunotherapeutic interventions. This review also explores various immunotherapeutic modalities, including immune checkpoint inhibitors (ICIs), adoptive cell therapies, and cancer vaccines, that leverage the DDR and neoantigen interplay to enhance anti-tumor immune responses. Moreover, the potential for developing vaccines targeting DDR-related neoantigens opens new frontiers in preventive and therapeutic strategies for TNBC. The rational design of vaccines tailored to the individual mutational landscape of TNBC holds promise for precision medicine approaches. In conclusion, the convergence of DDR and neoantigens in TNBC presents a compelling rationale for the development of innovative immunotherapies and vaccines. Understanding and targeting these interconnected processes may pave the way for personalized and effective interventions, offering new hope for patients grappling with the challenges posed by TNBCs.

三阴性乳腺癌(TNBC)因其侵袭性强、治疗方案有限而成为一项重大的临床挑战。DNA损伤应答(DDR)机制与新抗原的出现之间的相互作用为开发针对TNBC的靶向免疫治疗策略和疫苗提供了一条前景广阔的途径。DDR 是一个复杂的细胞机制网络,旨在维持基因组的完整性。在以遗传不稳定性为特征的 TNBC 中,DDR 成分的失调在肿瘤发生和发展中起着关键作用。本综述探讨了 DDR 与新抗原之间错综复杂的关系,揭示了 TNBC 细胞的潜在脆弱性。新抗原产生于癌细胞中的体细胞突变,是免疫系统可以识别的独特抗原。TNBC 的基因组不稳定性导致突变负荷增加,从而产生了丰富的新抗原。TNBC 中 DDR 和新抗原的融合为免疫疗法的靶向治疗提供了独特的机会。免疫疗法利用免疫系统选择性地靶向癌细胞,从而彻底改变了癌症治疗。TNBC 中与 DDR 相关的新抗原所具有的独特免疫原性使其成为免疫治疗干预的理想靶点。本综述还探讨了各种免疫治疗模式,包括免疫检查点抑制剂 (ICI)、收养细胞疗法和癌症疫苗,它们利用 DDR 和新抗原的相互作用来增强抗肿瘤免疫反应。此外,针对 DDR 相关新抗原开发疫苗的潜力为 TNBC 的预防和治疗策略开辟了新的领域。根据 TNBC 的个体突变情况合理设计疫苗为精准医疗方法带来了希望。总之,TNBC 中 DDR 和新抗原的融合为开发创新型免疫疗法和疫苗提供了令人信服的理由。了解并针对这些相互关联的过程可能会为个性化的有效干预铺平道路,为患者应对 TNBC 带来的挑战带来新的希望。
{"title":"DNA damage response and neoantigens: A favorable target for triple-negative breast cancer immunotherapy and vaccine development.","authors":"Rajasekaran Subbarayan, Dhasarathdev Srinivasan, Ranjith Balakrishnan, Ajeet Kumar, Salman Sadullah Usmani, Nityanand Srivastava","doi":"10.1016/bs.ircmb.2024.05.001","DOIUrl":"https://doi.org/10.1016/bs.ircmb.2024.05.001","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) poses a significant clinical challenge due to its aggressive nature and limited therapeutic options. The interplay between DNA damage response (DDR) mechanisms and the emergence of neoantigens represents a promising avenue for developing targeted immunotherapeutic strategies and vaccines for TNBC. The DDR is a complex network of cellular mechanisms designed to maintain genomic integrity. In TNBC, where genetic instability is a hallmark, dysregulation of DDR components plays a pivotal role in tumorigenesis and progression. This review explores the intricate relationship between DDR and neoantigens, shedding light on the potential vulnerabilities of TNBC cells. Neoantigens, arising from somatic mutations in cancer cells, represent unique antigens that can be recognized by the immune system. TNBC's propensity for genomic instability leads to an increased mutational burden, consequently yielding a rich repertoire of neoantigens. The convergence of DDR and neoantigens in TNBC offers a distinctive opportunity for immunotherapeutic targeting. Immunotherapy has revolutionized cancer treatment by harnessing the immune system to selectively target cancer cells. The unique immunogenicity conferred by DDR-related neoantigens in TNBC positions them as ideal targets for immunotherapeutic interventions. This review also explores various immunotherapeutic modalities, including immune checkpoint inhibitors (ICIs), adoptive cell therapies, and cancer vaccines, that leverage the DDR and neoantigen interplay to enhance anti-tumor immune responses. Moreover, the potential for developing vaccines targeting DDR-related neoantigens opens new frontiers in preventive and therapeutic strategies for TNBC. The rational design of vaccines tailored to the individual mutational landscape of TNBC holds promise for precision medicine approaches. In conclusion, the convergence of DDR and neoantigens in TNBC presents a compelling rationale for the development of innovative immunotherapies and vaccines. Understanding and targeting these interconnected processes may pave the way for personalized and effective interventions, offering new hope for patients grappling with the challenges posed by TNBCs.</p>","PeriodicalId":14422,"journal":{"name":"International review of cell and molecular biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142465574","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Regulation of lipid and serine metabolism by the oncogene c-Myc. 癌基因 c-Myc 对脂质和丝氨酸代谢的调控
3区 生物学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-01 Epub Date: 2024-03-27 DOI: 10.1016/bs.ircmb.2024.03.005
Subhajit Chatterjee, Prarthana Prashanth, Vipin Rawat, Sounak Ghosh Roy

Tumor formation is supported by metabolic reprogramming, characterized by increase nutrient uptake, glycolysis and glutaminolysis. The c-Myc proto-oncogene is a transcription factor, upregulated in most cancers and several reports showed the role of c-Myc in other metabolic pathways such as glucose, amino acid, and nucleotide metabolism. In this short report, we tried to summarize the existing takeaway points from studies conducted in different cancer types with respect to c-Myc and lipid and serine metabolism. Here, we report that c-Myc can activate both lipid and serine metabolism against the backdrop of tumor formation, and different therapies like aspirin and lomitapide target the links between c-Myc and metabolism to slow down tumor progression and invasion. We also report diverse upstream regulators that influence c-Myc in different cancers, and interestingly components of the lipid metabolism (like lipid phosphate phosphatase and leptin) and serine metabolism can also act upstream of c-Myc in certain occasions. Finally, we also summarize the existing knowledge on the involvement of epigenetic pathways and non-coding RNAs in regulating lipid and serine metabolism and c-Myc in tumor cells. Identification of non-coding factors and epigenetic mechanisms present a promising avenue of study that could empower researchers with novel anticancer treatment targeting c-Myc and lipid and serine metabolism pathways!

肿瘤的形成有赖于新陈代谢的重编程,其特点是营养摄取、糖酵解和谷氨酰胺酵解的增加。c-Myc 原癌基因是一种转录因子,在大多数癌症中都会上调,一些报告显示了 c-Myc 在葡萄糖、氨基酸和核苷酸代谢等其他代谢途径中的作用。在这份简短的报告中,我们试图总结在不同癌症类型中进行的有关 c-Myc 与脂质和丝氨酸代谢研究的现有要点。在此,我们报告了 c-Myc 可在肿瘤形成的背景下激活脂质和丝氨酸代谢,阿司匹林和洛米他匹等不同疗法可针对 c-Myc 与代谢之间的联系减缓肿瘤的进展和侵袭。我们还报告了在不同癌症中影响 c-Myc 的各种上游调节因子,有趣的是,脂质代谢(如脂质磷酸磷酸酶和瘦素)和丝氨酸代谢的成分在某些情况下也会作用于 c-Myc 的上游。最后,我们还总结了有关表观遗传途径和非编码 RNA 参与调控肿瘤细胞脂质和丝氨酸代谢以及 c-Myc 的现有知识。非编码因子和表观遗传机制的鉴定为研究人员提供了一条前景广阔的研究途径,可帮助他们针对 c-Myc 以及脂质和丝氨酸代谢途径进行新型抗癌治疗!
{"title":"Regulation of lipid and serine metabolism by the oncogene c-Myc.","authors":"Subhajit Chatterjee, Prarthana Prashanth, Vipin Rawat, Sounak Ghosh Roy","doi":"10.1016/bs.ircmb.2024.03.005","DOIUrl":"10.1016/bs.ircmb.2024.03.005","url":null,"abstract":"<p><p>Tumor formation is supported by metabolic reprogramming, characterized by increase nutrient uptake, glycolysis and glutaminolysis. The c-Myc proto-oncogene is a transcription factor, upregulated in most cancers and several reports showed the role of c-Myc in other metabolic pathways such as glucose, amino acid, and nucleotide metabolism. In this short report, we tried to summarize the existing takeaway points from studies conducted in different cancer types with respect to c-Myc and lipid and serine metabolism. Here, we report that c-Myc can activate both lipid and serine metabolism against the backdrop of tumor formation, and different therapies like aspirin and lomitapide target the links between c-Myc and metabolism to slow down tumor progression and invasion. We also report diverse upstream regulators that influence c-Myc in different cancers, and interestingly components of the lipid metabolism (like lipid phosphate phosphatase and leptin) and serine metabolism can also act upstream of c-Myc in certain occasions. Finally, we also summarize the existing knowledge on the involvement of epigenetic pathways and non-coding RNAs in regulating lipid and serine metabolism and c-Myc in tumor cells. Identification of non-coding factors and epigenetic mechanisms present a promising avenue of study that could empower researchers with novel anticancer treatment targeting c-Myc and lipid and serine metabolism pathways!</p>","PeriodicalId":14422,"journal":{"name":"International review of cell and molecular biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142465577","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epigenetic contributions to cancer: Exploring the role of glycation reactions. 表观遗传对癌症的影响:探索糖化反应的作用
3区 生物学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-01 Epub Date: 2024-05-31 DOI: 10.1016/bs.ircmb.2024.04.001
Hamda Khan, Zeeshan Rafi, Mohd Yasir Khan, Farah Maarfi, Shahnawaz Rehman, Kirtanjot Kaur, Mohammad Kaleem Ahmad, Uzma Shahab, Naved Ahmad, Saheem Ahmad

Advanced Glycation End-products (AGEs), with their prolonged half-life in the human body, are emerging as potent diagnostic indicators. Early intervention studies, focusing on AGE cross-link breakers, have shown encouraging results in heart failure patients, paving the way for disease progression monitoring and therapy effectiveness evaluation. AGEs are the byproducts of a non-enzymatic reaction where sugars interact with proteins, lipids, and nucleic acids. These compounds possess the power to alter numerous biological processes, ranging from disrupting molecular conformation and promoting cross-linking to modifying enzyme activity, reducing clearance, and impairing receptor recognition. The damage inflicted by AGEs through the stimulation of intracellular signaling pathways is associated with the onset of chronic diseases across various organ systems. This review consolidates the characteristics of AGEs and the challenges posed by their expression in diverse physiological and pathological states. Furthermore, it highlights the clinical relevance of AGEs and the latest research breakthroughs aimed at reducing AGE accumulation.

高级糖化终产物(AGEs)在人体内的半衰期较长,正在成为有效的诊断指标。以 AGE 交联断裂物为重点的早期干预研究在心衰患者中取得了令人鼓舞的结果,为疾病进展监测和疗效评估铺平了道路。AGE 是糖与蛋白质、脂类和核酸相互作用的非酶反应的副产品。这些化合物能够改变许多生物过程,包括破坏分子构象、促进交联、改变酶活性、降低清除率和损害受体识别等。AGEs 通过刺激细胞内信号通路造成的损害与各器官系统慢性疾病的发病有关。本综述总结了 AGEs 的特点及其在不同生理和病理状态下的表达所带来的挑战。此外,它还强调了 AGEs 的临床意义以及旨在减少 AGE 积累的最新研究突破。
{"title":"Epigenetic contributions to cancer: Exploring the role of glycation reactions.","authors":"Hamda Khan, Zeeshan Rafi, Mohd Yasir Khan, Farah Maarfi, Shahnawaz Rehman, Kirtanjot Kaur, Mohammad Kaleem Ahmad, Uzma Shahab, Naved Ahmad, Saheem Ahmad","doi":"10.1016/bs.ircmb.2024.04.001","DOIUrl":"https://doi.org/10.1016/bs.ircmb.2024.04.001","url":null,"abstract":"<p><p>Advanced Glycation End-products (AGEs), with their prolonged half-life in the human body, are emerging as potent diagnostic indicators. Early intervention studies, focusing on AGE cross-link breakers, have shown encouraging results in heart failure patients, paving the way for disease progression monitoring and therapy effectiveness evaluation. AGEs are the byproducts of a non-enzymatic reaction where sugars interact with proteins, lipids, and nucleic acids. These compounds possess the power to alter numerous biological processes, ranging from disrupting molecular conformation and promoting cross-linking to modifying enzyme activity, reducing clearance, and impairing receptor recognition. The damage inflicted by AGEs through the stimulation of intracellular signaling pathways is associated with the onset of chronic diseases across various organ systems. This review consolidates the characteristics of AGEs and the challenges posed by their expression in diverse physiological and pathological states. Furthermore, it highlights the clinical relevance of AGEs and the latest research breakthroughs aimed at reducing AGE accumulation.</p>","PeriodicalId":14422,"journal":{"name":"International review of cell and molecular biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142046654","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Factors affecting heterogeneity in breast cancer microenvironment: A narrative mini review. 影响乳腺癌微环境异质性的因素:叙事性微型综述。
3区 生物学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-01 Epub Date: 2024-02-15 DOI: 10.1016/bs.ircmb.2024.01.002
Anirban Ganguly, Sumit Mukherjee, Kaushiki Chatterjee, Sheila Spada

Breast cancer (BC) heterogeneity is a key trait of BC tumors with crucial implications on tumorigenesis, diagnosis, and therapeutic modalities. It is influenced by tumor intrinsic features and by the tumor microenvironment (TME) composition of different intra-tumoral regions, which in turn affect cancer progression within patients. In this mini review, we will highlight the mechanisms that generate cancer heterogeneity in BC and how they affect the responses to cancer therapies.

乳腺癌(BC)的异质性是BC肿瘤的一个关键特征,对肿瘤发生、诊断和治疗方法具有重要影响。异质性受肿瘤内在特征和不同瘤内区域肿瘤微环境(TME)组成的影响,进而影响患者的癌症进展。在这篇小型综述中,我们将重点介绍 BC 癌症异质性的产生机制,以及它们如何影响癌症疗法的反应。
{"title":"Factors affecting heterogeneity in breast cancer microenvironment: A narrative mini review.","authors":"Anirban Ganguly, Sumit Mukherjee, Kaushiki Chatterjee, Sheila Spada","doi":"10.1016/bs.ircmb.2024.01.002","DOIUrl":"10.1016/bs.ircmb.2024.01.002","url":null,"abstract":"<p><p>Breast cancer (BC) heterogeneity is a key trait of BC tumors with crucial implications on tumorigenesis, diagnosis, and therapeutic modalities. It is influenced by tumor intrinsic features and by the tumor microenvironment (TME) composition of different intra-tumoral regions, which in turn affect cancer progression within patients. In this mini review, we will highlight the mechanisms that generate cancer heterogeneity in BC and how they affect the responses to cancer therapies.</p>","PeriodicalId":14422,"journal":{"name":"International review of cell and molecular biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140856400","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting signaling pathways in cancer stem cells: A potential approach for developing novel anti-cancer therapeutics. 瞄准癌症干细胞的信号通路:开发新型抗癌疗法的潜在方法。
3区 生物学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-01-01 Epub Date: 2024-02-10 DOI: 10.1016/bs.ircmb.2024.01.001
Saptarshi Sinha, Krushna Chandra Hembram, Subhajit Chatterjee

Cancer stem cells (CSCs) have emerged as prime players in the intricate landscape of cancer development, progression, and resistance to traditional treatments. These unique cellular subpopulations own the remarkable capability of self-renewal and differentiation, giving rise to the diverse cellular makeup of tumors and fostering their recurrence following conventional therapies. In the quest for developing more effective cancer therapeutics, the focus has now shifted toward targeting the signaling pathways that govern CSCs behavior. This chapter underscores the significance of these signaling pathways in CSC biology and their potential as pivotal targets for the development of novel chemotherapy approaches. We delve into several key signaling pathways essential for maintaining the defining characteristics of CSCs, including the Wnt, Hedgehog, Notch, JAK-STAT, NF-κB pathways, among others, shedding light on their potential crosstalk. Furthermore, we highlight the latest advancements in CSC-targeted therapies, spanning from promising preclinical models to ongoing clinical trials. A comprehensive understanding of the intricate molecular aspects of CSC signaling pathways and their manipulation holds the prospective to revolutionize cancer treatment paradigms. This, in turn, could lead to more efficacious and personalized therapies with the ultimate goal of eradicating CSCs and enhancing overall patient outcomes. The exploration of CSC signaling pathways represents a key step towards a brighter future in the battle against cancer.

癌症干细胞(CSCs)已成为癌症发展、恶化和对传统疗法产生抗药性等错综复杂问题的主要参与者。这些独特的细胞亚群具有非凡的自我更新和分化能力,形成了肿瘤的多种细胞构成,并在传统疗法后促使肿瘤复发。为了开发更有效的癌症疗法,目前的研究重点已转向靶向调节 CSCs 行为的信号通路。本章强调了这些信号通路在 CSC 生物学中的重要性,以及它们作为开发新型化疗方法关键靶点的潜力。我们深入探讨了维持 CSCs 决定性特征所必需的几种关键信号通路,包括 Wnt、刺猬、Notch、JAK-STAT、NF-κB 通路等,并揭示了它们之间潜在的相互影响。此外,我们还重点介绍了干细胞靶向疗法的最新进展,包括从前景看好的临床前模型到正在进行的临床试验。全面了解 CSC 信号通路错综复杂的分子方面及其操作方法有望彻底改变癌症治疗模式。这反过来又会带来更有效的个性化疗法,最终目标是消灭 CSC 并改善患者的整体预后。探索 CSC 信号通路是迈向抗癌斗争光明未来的关键一步。
{"title":"Targeting signaling pathways in cancer stem cells: A potential approach for developing novel anti-cancer therapeutics.","authors":"Saptarshi Sinha, Krushna Chandra Hembram, Subhajit Chatterjee","doi":"10.1016/bs.ircmb.2024.01.001","DOIUrl":"10.1016/bs.ircmb.2024.01.001","url":null,"abstract":"<p><p>Cancer stem cells (CSCs) have emerged as prime players in the intricate landscape of cancer development, progression, and resistance to traditional treatments. These unique cellular subpopulations own the remarkable capability of self-renewal and differentiation, giving rise to the diverse cellular makeup of tumors and fostering their recurrence following conventional therapies. In the quest for developing more effective cancer therapeutics, the focus has now shifted toward targeting the signaling pathways that govern CSCs behavior. This chapter underscores the significance of these signaling pathways in CSC biology and their potential as pivotal targets for the development of novel chemotherapy approaches. We delve into several key signaling pathways essential for maintaining the defining characteristics of CSCs, including the Wnt, Hedgehog, Notch, JAK-STAT, NF-κB pathways, among others, shedding light on their potential crosstalk. Furthermore, we highlight the latest advancements in CSC-targeted therapies, spanning from promising preclinical models to ongoing clinical trials. A comprehensive understanding of the intricate molecular aspects of CSC signaling pathways and their manipulation holds the prospective to revolutionize cancer treatment paradigms. This, in turn, could lead to more efficacious and personalized therapies with the ultimate goal of eradicating CSCs and enhancing overall patient outcomes. The exploration of CSC signaling pathways represents a key step towards a brighter future in the battle against cancer.</p>","PeriodicalId":14422,"journal":{"name":"International review of cell and molecular biology","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140864743","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"生物学","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
International review of cell and molecular biology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1