首页 > 最新文献

癌症耐药(英文)最新文献

英文 中文
Mechanisms of immunotherapy resistance in small cell lung cancer. 小细胞肺癌免疫治疗耐药机制。
IF 4.6 Q1 ONCOLOGY Pub Date : 2024-12-28 eCollection Date: 2024-01-01 DOI: 10.20517/cdr.2024.154
Yunan Nie, Kurt A Schalper, Anne Chiang

Small-cell lung cancer (SCLC) is an aggressive neuroendocrine tumor with a poor prognosis. Although the addition of immunotherapy to chemotherapy has modestly improved outcomes, most patients rapidly develop resistance. Resistance to immunotherapy can be broadly categorized into primary resistance and acquired resistance, as proposed by the Society for Immunotherapy of Cancer (SITC) consensus definition. Primary resistance occurs in the setting of failure to respond to immune checkpoint inhibitors (ICIs), while acquired resistance develops after initial response. The mechanisms of acquired and primary resistance to ICI are not well understood in SCLC, denoting an area of critical unmet need. Both intrinsic and extrinsic mechanisms play significant roles in immunotherapy resistance. Intrinsic mechanisms include defects in antigen presentation, mutations in key genes, reduced tumor immunogenicity, and epigenetic alterations. Extrinsic mechanisms involve the tumor microenvironment (TME), which is a complex interplay of both tumor- and immunosuppressive immune cells, vasculature, and microbiome. An understanding of these resistance mechanisms is crucial for developing novel therapeutic strategies to advance effective immunotherapy in patients with SCLC, a critical area of unmet need.

小细胞肺癌是一种侵袭性神经内分泌肿瘤,预后较差。虽然在化疗的基础上加入免疫疗法有一定程度的改善,但大多数患者会迅速产生耐药性。根据癌症免疫治疗学会(Society for immunotherapy of Cancer, SITC)共识定义,免疫治疗耐药大致可分为原发性耐药和获得性耐药。原发性耐药发生在对免疫检查点抑制剂(ICIs)反应失败的情况下,而获得性耐药发生在初始反应之后。在SCLC中,对ICI的获得性和原发性耐药机制尚不清楚,这表明一个关键的未满足需求的领域。免疫治疗耐药的内在机制和外在机制都起着重要作用。内在机制包括抗原呈递缺陷、关键基因突变、肿瘤免疫原性降低和表观遗传改变。外部机制涉及肿瘤微环境(TME),这是肿瘤和免疫抑制免疫细胞、脉管系统和微生物组的复杂相互作用。了解这些耐药机制对于开发新的治疗策略以推进SCLC患者的有效免疫治疗至关重要,这是一个未满足需求的关键领域。
{"title":"Mechanisms of immunotherapy resistance in small cell lung cancer.","authors":"Yunan Nie, Kurt A Schalper, Anne Chiang","doi":"10.20517/cdr.2024.154","DOIUrl":"10.20517/cdr.2024.154","url":null,"abstract":"<p><p>Small-cell lung cancer (SCLC) is an aggressive neuroendocrine tumor with a poor prognosis. Although the addition of immunotherapy to chemotherapy has modestly improved outcomes, most patients rapidly develop resistance. Resistance to immunotherapy can be broadly categorized into primary resistance and acquired resistance, as proposed by the Society for Immunotherapy of Cancer (SITC) consensus definition. Primary resistance occurs in the setting of failure to respond to immune checkpoint inhibitors (ICIs), while acquired resistance develops after initial response. The mechanisms of acquired and primary resistance to ICI are not well understood in SCLC, denoting an area of critical unmet need. Both intrinsic and extrinsic mechanisms play significant roles in immunotherapy resistance. Intrinsic mechanisms include defects in antigen presentation, mutations in key genes, reduced tumor immunogenicity, and epigenetic alterations. Extrinsic mechanisms involve the tumor microenvironment (TME), which is a complex interplay of both tumor- and immunosuppressive immune cells, vasculature, and microbiome. An understanding of these resistance mechanisms is crucial for developing novel therapeutic strategies to advance effective immunotherapy in patients with SCLC, a critical area of unmet need.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"7 ","pages":"55"},"PeriodicalIF":4.6,"publicationDate":"2024-12-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11724353/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142973507","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mitochondrial genome variability and metabolic alterations reveal new biomarkers of resistance in testicular germ cell tumors. 线粒体基因组变异和代谢改变揭示了睾丸生殖细胞肿瘤耐药的新生物标志物。
IF 4.6 Q1 ONCOLOGY Pub Date : 2024-12-18 eCollection Date: 2024-01-01 DOI: 10.20517/cdr.2024.141
Pavlina Kabelikova, Danica Ivovic, Zuzana Sumbalova, Miloslav Karhanek, Lucia Tatayova, Martina Skopkova, Michal Cagalinec, Vladimira Bruderova, Jan Roska, Dana Jurkovicova

Aim: Mutations in the mitochondrial (mt) genome contribute to metabolic dysfunction and their accumulation relates to disease progression and resistance development in cancer cells. This study explores the mutational status of the mt genome of cisplatin-resistant vs. -sensitive testicular germ cell tumor (TGCT) cells and explores its association with their respiration parameters, expression of respiratory genes, and preferences for metabolic pathways to reveal new markers of therapy resistance in TGCTs. Methods: Using Illumina sequencing with Twist Enrichment Panel, the mutations of mt genomes of sensitive 2102EP, H12.1, NTERA-2, T-cam and resistant 2102EP Cis, H12.1 ODM, 1411HP, 1777NRpmet, NTERA-2 Cis and T-cam Cis cell lines were identified. The mt respiration of the cells was assessed using high-resolution respirometry method (O2k-respirometer Oroboros) and the differential expression profiles of mt respiratory genes were determined using RT-qPCR. Associated preferences for metabolic pathways were compared using Glycolysis/OXPHOS assay. Results: In resistant TGCT cells, new mutations in mt genes MT-ND1-6, MT-RNR, MT-CO1-3, MT-ATP6, and MT-CYB were recognized. The respiratory rates of the 1777NRpmet cell line were the highest, while those of the 1411HP line the lowest; rates of the control and all other TGCT cell lines fell between these two lines. The statistically significant differences in gene expression of the respiratory genes were recorded only in NTERA-2 Cis and T-cam Cis cell lines. Sensitive cell lines NTERA-2 and 2102EP preferred oxidative phosphorylation (OXPHOS), while glycolysis was typical for resistant NTERA-2 Cis, 2102EP Cis and 1411HP cell lines. Metastatic 1777NRpmet cells seem to utilize both. An isogenic pair of cell lines H12.1 and H12.1ODM showed the opposite dependence, sensitive H12.1 preferring glycolysis, while resistant H12.1ODM OXPHOS. Conclusion: In summary, our study identified new mutations in mt genes of resistant TGCT cell lines that are associated with different mt respiration parameters, gene expression patterns and preferences for metabolic pathways, providing potential novel molecular biomarkers that distinguish the resistant TGCT phenotype or specify its histological classification.

目的:线粒体(mt)基因组突变会导致代谢功能障碍,其积累与癌细胞的疾病进展和抗药性发展有关。本研究探讨了顺铂耐药与敏感睾丸生殖细胞瘤(TGCT)细胞线粒体基因组的突变状态,并探讨了其与呼吸参数、呼吸基因表达和代谢途径偏好的关联,以揭示 TGCT 治疗耐药性的新标记。研究方法利用Illumina测序技术和Twist Enrichment Panel,鉴定了敏感的2102EP、H12.1、NTERA-2、T-cam和耐药的2102EP Cis、H12.1 ODM、1411HP、1777NRpmet、NTERA-2 Cis和T-cam Cis细胞系的mt基因组突变。使用高分辨率呼吸测定法(O2k-respirometer Oroboros)评估了细胞的黑质呼吸,并使用 RT-qPCR 确定了黑质呼吸基因的差异表达谱。使用糖酵解/OXPHOS 检测比较了代谢途径的相关偏好。结果:在抗性 TGCT 细胞中,发现了 MT-ND1-6、MT-RNR、MT-CO1-3、MT-ATP6 和 MT-CYB 等 mt 基因的新突变。1777NRpmet 细胞系的呼吸率最高,而 1411HP 细胞系的呼吸率最低;对照组和所有其他 TGCT 细胞系的呼吸率介于这两个细胞系之间。只有 NTERA-2 顺式细胞系和 T-cam 顺式细胞系的呼吸基因表达有明显的统计学差异。敏感细胞株 NTERA-2 和 2102EP 更倾向于氧化磷酸化(OXPHOS),而耐药细胞株 NTERA-2 Cis、2102EP Cis 和 1411HP 则以糖酵解为典型特征。转移性 1777NRpmet 细胞似乎同时利用这两种方式。H12.1和H12.1ODM这对同源细胞株显示出相反的依赖性,敏感的H12.1更倾向于糖酵解,而耐药的H12.1ODM则是OXPHOS。结论总之,我们的研究发现了耐药 TGCT 细胞系 mt 基因的新突变,这些突变与不同的 mt 呼吸参数、基因表达模式和代谢途径偏好有关,为区分耐药 TGCT 表型或明确其组织学分类提供了潜在的新型分子生物标记物。
{"title":"Mitochondrial genome variability and metabolic alterations reveal new biomarkers of resistance in testicular germ cell tumors.","authors":"Pavlina Kabelikova, Danica Ivovic, Zuzana Sumbalova, Miloslav Karhanek, Lucia Tatayova, Martina Skopkova, Michal Cagalinec, Vladimira Bruderova, Jan Roska, Dana Jurkovicova","doi":"10.20517/cdr.2024.141","DOIUrl":"10.20517/cdr.2024.141","url":null,"abstract":"<p><p><b>Aim:</b> Mutations in the mitochondrial (mt) genome contribute to metabolic dysfunction and their accumulation relates to disease progression and resistance development in cancer cells. This study explores the mutational status of the mt genome of cisplatin-resistant <i>vs.</i> -sensitive testicular germ cell tumor (TGCT) cells and explores its association with their respiration parameters, expression of respiratory genes, and preferences for metabolic pathways to reveal new markers of therapy resistance in TGCTs. <b>Methods:</b> Using Illumina sequencing with Twist Enrichment Panel, the mutations of mt genomes of sensitive 2102EP, H12.1, NTERA-2, T-cam and resistant 2102EP Cis, H12.1 ODM, 1411HP, 1777NRpmet, NTERA-2 Cis and T-cam Cis cell lines were identified. The mt respiration of the cells was assessed using high-resolution respirometry method (O2k-respirometer Oroboros) and the differential expression profiles of mt respiratory genes were determined using RT-qPCR. Associated preferences for metabolic pathways were compared using Glycolysis/OXPHOS assay. <b>Results:</b> In resistant TGCT cells, new mutations in mt genes <i>MT-ND1-6, MT-RNR, MT-CO1-3, MT-ATP6</i>, and <i>MT-CYB</i> were recognized. The respiratory rates of the 1777NRpmet cell line were the highest, while those of the 1411HP line the lowest; rates of the control and all other TGCT cell lines fell between these two lines. The statistically significant differences in gene expression of the respiratory genes were recorded only in NTERA-2 Cis and T-cam Cis cell lines. Sensitive cell lines NTERA-2 and 2102EP preferred oxidative phosphorylation (OXPHOS), while glycolysis was typical for resistant NTERA-2 Cis, 2102EP Cis and 1411HP cell lines. Metastatic 1777NRpmet cells seem to utilize both. An isogenic pair of cell lines H12.1 and H12.1ODM showed the opposite dependence, sensitive H12.1 preferring glycolysis, while resistant H12.1ODM OXPHOS. <b>Conclusion:</b> In summary, our study identified new mutations in mt genes of resistant TGCT cell lines that are associated with different mt respiration parameters, gene expression patterns and preferences for metabolic pathways, providing potential novel molecular biomarkers that distinguish the resistant TGCT phenotype or specify its histological classification.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"7 ","pages":"54"},"PeriodicalIF":4.6,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11724352/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142973523","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ovarian tumor microenvironment contributes to tumor progression and chemoresistance. 卵巢肿瘤微环境与肿瘤进展及化疗耐药有关。
IF 4.6 Q1 ONCOLOGY Pub Date : 2024-12-17 eCollection Date: 2024-01-01 DOI: 10.20517/cdr.2024.111
Adriana Ponton-Almodovar, Samuel Sanderson, Ramandeep Rattan, Jamie J Bernard, Sachi Horibata

Ovarian cancer is one of the deadliest gynecologic cancers affecting the female reproductive tract. This is largely attributed to frequent recurrence and development of resistance to the platinum-based drugs cisplatin and carboplatin. One of the major contributing factors to increased cancer progression and resistance to chemotherapy is the tumor microenvironment (TME). Extracellular signaling from cells within the microenvironment heavily influences progression and drug resistance in ovarian cancer. This is frequently done through metabolic reprogramming, the process where cancer cells switch between biochemical pathways to increase their chances of survival and proliferation. Here, we focus on how crosstalk between components of the TME and the tumor promotes resistance to platinum-based chemotherapy. We highlight the role of cancer-associated fibroblasts, immune cells, adipocytes, and endothelial cells in ovarian tumor progression, invasion, metastasis, and chemoresistance. We also highlight recent advancements in targeting components of the TME as a novel therapeutic avenue to combat chemoresistance in ovarian cancer.

卵巢癌是影响女性生殖道的最致命的妇科癌症之一。这主要是由于对铂类药物顺铂和卡铂的频繁复发和耐药性的发展。肿瘤微环境(tumor microenvironment, TME)是导致肿瘤进展加快和化疗耐药的主要因素之一。微环境中细胞发出的细胞外信号严重影响卵巢癌的进展和耐药性。这通常是通过代谢重编程来实现的,在这个过程中,癌细胞在生化途径之间切换,以增加它们生存和增殖的机会。在这里,我们关注的是TME和肿瘤组分之间的串扰如何促进对铂基化疗的耐药性。我们强调癌症相关成纤维细胞、免疫细胞、脂肪细胞和内皮细胞在卵巢肿瘤进展、侵袭、转移和化疗耐药中的作用。我们还强调了靶向TME成分作为对抗卵巢癌化疗耐药的新治疗途径的最新进展。
{"title":"Ovarian tumor microenvironment contributes to tumor progression and chemoresistance.","authors":"Adriana Ponton-Almodovar, Samuel Sanderson, Ramandeep Rattan, Jamie J Bernard, Sachi Horibata","doi":"10.20517/cdr.2024.111","DOIUrl":"10.20517/cdr.2024.111","url":null,"abstract":"<p><p>Ovarian cancer is one of the deadliest gynecologic cancers affecting the female reproductive tract. This is largely attributed to frequent recurrence and development of resistance to the platinum-based drugs cisplatin and carboplatin. One of the major contributing factors to increased cancer progression and resistance to chemotherapy is the tumor microenvironment (TME). Extracellular signaling from cells within the microenvironment heavily influences progression and drug resistance in ovarian cancer. This is frequently done through metabolic reprogramming, the process where cancer cells switch between biochemical pathways to increase their chances of survival and proliferation. Here, we focus on how crosstalk between components of the TME and the tumor promotes resistance to platinum-based chemotherapy. We highlight the role of cancer-associated fibroblasts, immune cells, adipocytes, and endothelial cells in ovarian tumor progression, invasion, metastasis, and chemoresistance. We also highlight recent advancements in targeting components of the TME as a novel therapeutic avenue to combat chemoresistance in ovarian cancer.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"7 ","pages":"53"},"PeriodicalIF":4.6,"publicationDate":"2024-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11724355/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142973544","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Role of the TME in immune checkpoint blockade resistance of non-small cell lung cancer. TME在非小细胞肺癌免疫检查点阻断抵抗中的作用。
IF 4.6 Q1 ONCOLOGY Pub Date : 2024-12-16 eCollection Date: 2024-01-01 DOI: 10.20517/cdr.2024.166
Yuening Dai, Xueqi Tian, Xuanting Ye, Yabin Gong, Ling Xu, Lijing Jiao

Primary and secondary resistance to immune checkpoint blockade (ICB) reduces its efficacy. The mechanisms underlying immunotherapy resistance are highly complex. In non-small cell lung cancer (NSCLC), these mechanisms are primarily associated with the loss of programmed cell death-ligand 1 (PD-L1) expression, genetic mutations, circular RNA axis and transcription factor regulation, antigen presentation disorders, and dysregulation of signaling pathways. Additionally, alterations in the tumor microenvironment (TME) play a pivotal role in driving immunotherapy resistance. Primary resistance is mainly attributed to TME alterations, including mutations and co-mutations, modulation of T cell infiltration, enrichment of M2 tumor-associated macrophages (M2-TAMs) and mucosal-associated invariant T (MAIT) cells, vascular endothelial growth factor (VEGF), and pulmonary fibrosis. Acquired resistance mainly stems from changes in cellular infiltration patterns leading to "cold" or "hot" tumors, altered interferon (IFN) signaling pathway expression, involvement of extracellular vesicles (EVs), and oxidative stress responses, as well as post-treatment gene mutations and circadian rhythm disruption (CRD). This review presents an overview of various mechanisms underlying resistance to ICB, elucidates the alterations in the TME during primary, adaptive, and acquired resistance, and discusses existing strategies for overcoming ICB resistance.

免疫检查点阻断疗法(ICB)的原发性和继发性抗药性会降低其疗效。免疫疗法产生耐药性的机制非常复杂。在非小细胞肺癌(NSCLC)中,这些机制主要与程序性细胞死亡配体 1(PD-L1)表达丧失、基因突变、环状 RNA 轴和转录因子调控、抗原呈递紊乱以及信号通路失调有关。此外,肿瘤微环境(TME)的改变也是导致免疫疗法耐药的关键因素。原发性耐药性主要归因于肿瘤微环境的改变,包括突变和共突变、T细胞浸润的调节、M2肿瘤相关巨噬细胞(M2-TAMs)和粘膜相关不变T细胞(MAIT)的富集、血管内皮生长因子(VEGF)和肺纤维化。获得性耐药性主要源于导致 "冷 "或 "热 "肿瘤的细胞浸润模式变化、干扰素(IFN)信号通路表达的改变、细胞外囊泡(EVs)的参与、氧化应激反应以及治疗后基因突变和昼夜节律紊乱(CRD)。本综述概述了对 ICB 产生耐药性的各种机制,阐明了原发性、适应性和获得性耐药性期间 TME 的变化,并讨论了克服 ICB 耐药性的现有策略。
{"title":"Role of the TME in immune checkpoint blockade resistance of non-small cell lung cancer.","authors":"Yuening Dai, Xueqi Tian, Xuanting Ye, Yabin Gong, Ling Xu, Lijing Jiao","doi":"10.20517/cdr.2024.166","DOIUrl":"10.20517/cdr.2024.166","url":null,"abstract":"<p><p>Primary and secondary resistance to immune checkpoint blockade (ICB) reduces its efficacy. The mechanisms underlying immunotherapy resistance are highly complex. In non-small cell lung cancer (NSCLC), these mechanisms are primarily associated with the loss of programmed cell death-ligand 1 (PD-L1) expression, genetic mutations, circular RNA axis and transcription factor regulation, antigen presentation disorders, and dysregulation of signaling pathways. Additionally, alterations in the tumor microenvironment (TME) play a pivotal role in driving immunotherapy resistance. Primary resistance is mainly attributed to TME alterations, including mutations and co-mutations, modulation of T cell infiltration, enrichment of M2 tumor-associated macrophages (M2-TAMs) and mucosal-associated invariant T (MAIT) cells, vascular endothelial growth factor (VEGF), and pulmonary fibrosis. Acquired resistance mainly stems from changes in cellular infiltration patterns leading to \"cold\" or \"hot\" tumors, altered interferon (IFN) signaling pathway expression, involvement of extracellular vesicles (EVs), and oxidative stress responses, as well as post-treatment gene mutations and circadian rhythm disruption (CRD). This review presents an overview of various mechanisms underlying resistance to ICB, elucidates the alterations in the TME during primary, adaptive, and acquired resistance, and discusses existing strategies for overcoming ICB resistance.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"7 ","pages":"52"},"PeriodicalIF":4.6,"publicationDate":"2024-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11724356/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142973546","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CIRCUS: CIRCUlating tumour cells in soft tissue Sarcoma - a short report. 马戏团:软组织肉瘤中的循环肿瘤细胞-一个简短的报告。
IF 4.6 Q1 ONCOLOGY Pub Date : 2024-12-13 eCollection Date: 2024-01-01 DOI: 10.20517/cdr.2024.149
Robin J Young, Joanna E Chowdry, Denis Cochonneau, Dominique Heymann

Aims: Circulating tumour cells (CTCs) can be detected in peripheral blood using their physical properties (increased size and less deformable than normal circulating blood cells) or using cell surface markers. The study of these CTCs should provide important insights into tumour biology, including mechanisms of drug resistance. We performed a pilot study (IRAS ID: 235459) to evaluate if CTCs could be isolated from peripheral blood samples collected from soft tissue sarcoma (STS) patients. Methods: We used a combined approach that first enriched samples for CTCs using a microfluidic cassette via ParosrtixTMPR1, and then sorted cells stained for vimentin and cytokeratin using the DEPArrayTM. The total circulating cell-free DNA (cfDNA) level was also analysed. Data were correlated with clinical parameters. Results: 13 patients were recruited to this study: 7 patients with localised disease and 6 patients with metastatic disease. CTCs exhibited a high heterogeneity based on their expression of mesenchymal and epithelial markers. There was no significant difference in the number of CTCs between patients with localised versus metastatic disease. We observed no correlation between CTC numbers and cfDNA; however, the number of CTCs did correlate with primary tumour size. Conclusion: The present study demonstrates the presence of CTCs in STS patients with localised and advanced disease. Further and larger studies are needed to characterise STS CTCs and to evaluate their prognostic significance.

目的:循环肿瘤细胞(ctc)可以通过其物理特性(比正常循环血细胞大小增加且不易变形)或使用细胞表面标记物在外周血中检测。对这些CTCs的研究应该为肿瘤生物学提供重要的见解,包括耐药机制。我们进行了一项试点研究(IRAS ID: 235459),以评估是否可以从软组织肉瘤(STS)患者的外周血样本中分离出ctc。方法:我们采用了一种联合方法,首先使用微流控盒通过ParosrtixTMPR1富集ctc样品,然后使用DEPArrayTM对染色的vimentin和细胞角蛋白进行分选。同时分析循环游离细胞DNA (cfDNA)水平。数据与临床参数相关。结果:本研究招募了13例患者,其中7例为局部疾病,6例为转移性疾病。基于间充质和上皮标记物的表达,ctc表现出高度的异质性。在局部和转移性疾病患者中,ctc的数量没有显著差异。我们观察到CTC数与cfDNA没有相关性;然而,ctc的数量确实与原发肿瘤的大小相关。结论:本研究证实了局部和晚期STS患者存在CTCs。需要进一步和更大规模的研究来确定STS ctc的特征并评估其预后意义。
{"title":"CIRCUS: CIRCUlating tumour cells in soft tissue Sarcoma - a short report.","authors":"Robin J Young, Joanna E Chowdry, Denis Cochonneau, Dominique Heymann","doi":"10.20517/cdr.2024.149","DOIUrl":"10.20517/cdr.2024.149","url":null,"abstract":"<p><p><b>Aims:</b> Circulating tumour cells (CTCs) can be detected in peripheral blood using their physical properties (increased size and less deformable than normal circulating blood cells) or using cell surface markers. The study of these CTCs should provide important insights into tumour biology, including mechanisms of drug resistance. We performed a pilot study (IRAS ID: 235459) to evaluate if CTCs could be isolated from peripheral blood samples collected from soft tissue sarcoma (STS) patients. <b>Methods:</b> We used a combined approach that first enriched samples for CTCs using a microfluidic cassette via Parosrtix<sup>TM</sup>PR1, and then sorted cells stained for vimentin and cytokeratin using the DEPArray<sup>TM</sup>. The total circulating cell-free DNA (cfDNA) level was also analysed. Data were correlated with clinical parameters. <b>Results:</b> 13 patients were recruited to this study: 7 patients with localised disease and 6 patients with metastatic disease. CTCs exhibited a high heterogeneity based on their expression of mesenchymal and epithelial markers. There was no significant difference in the number of CTCs between patients with localised versus metastatic disease. We observed no correlation between CTC numbers and cfDNA; however, the number of CTCs did correlate with primary tumour size. <b>Conclusion:</b> The present study demonstrates the presence of CTCs in STS patients with localised and advanced disease. Further and larger studies are needed to characterise STS CTCs and to evaluate their prognostic significance.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"7 ","pages":"51"},"PeriodicalIF":4.6,"publicationDate":"2024-12-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11724351/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142973342","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Overcoming drug resistance through extracellular vesicle-based drug delivery system in cancer treatment. 基于细胞外囊泡的给药系统在癌症治疗中克服耐药性。
IF 4.6 Q1 ONCOLOGY Pub Date : 2024-12-12 eCollection Date: 2024-01-01 DOI: 10.20517/cdr.2024.107
Long Zheng, Ruibai Chang, Bingjing Liang, Yitong Wang, Yushan Zhu, Zijing Jia, Jindian Fan, Zhe Zhang, Bo Du, Dexin Kong

Drug resistance is a major challenge in cancer therapy that often leads to treatment failure and disease relapse. Despite advancements in chemotherapeutic agents and targeted therapies, cancers often develop drug resistance, making these treatments ineffective. Extracellular vesicles (EVs) have gained attention for their potential applications in drug delivery because of their natural origin, biocompatibility, and ability to cross biological barriers. Using the unique properties of EVs could enhance drug accumulation at target sites, minimize systemic toxicity, and precisely target specific cells. Here, we discuss the characteristics and functionalization of EVs, the mechanisms of drug resistance, and the applications of engineered EVs to overcome drug resistance. This review provides a comprehensive overview of the advancements in EV-based drug delivery systems and their applications in overcoming cancer drug resistance. We highlight the potential of EV-based drug delivery systems to revolutionize cancer therapy and offer promising strategies for more effective treatment modalities.

耐药性是癌症治疗中的一大挑战,常常导致治疗失败和疾病复发。尽管化疗药物和靶向疗法取得了进步,但癌症往往会产生耐药性,使这些治疗失效。细胞外囊泡(EVs)因其天然来源、生物相容性和穿越生物屏障的能力,在药物递送方面的潜在应用备受关注。利用细胞外囊泡的独特特性,可以提高药物在目标部位的蓄积,最大限度地减少全身毒性,并精确地靶向特定细胞。在此,我们将讨论 EVs 的特性和功能化、耐药机制以及工程化 EVs 在克服耐药性方面的应用。本综述全面概述了基于 EV 的给药系统的进展及其在克服癌症耐药性方面的应用。我们强调了基于 EV 的给药系统彻底改变癌症治疗的潜力,并为更有效的治疗模式提供了前景广阔的策略。
{"title":"Overcoming drug resistance through extracellular vesicle-based drug delivery system in cancer treatment.","authors":"Long Zheng, Ruibai Chang, Bingjing Liang, Yitong Wang, Yushan Zhu, Zijing Jia, Jindian Fan, Zhe Zhang, Bo Du, Dexin Kong","doi":"10.20517/cdr.2024.107","DOIUrl":"10.20517/cdr.2024.107","url":null,"abstract":"<p><p>Drug resistance is a major challenge in cancer therapy that often leads to treatment failure and disease relapse. Despite advancements in chemotherapeutic agents and targeted therapies, cancers often develop drug resistance, making these treatments ineffective. Extracellular vesicles (EVs) have gained attention for their potential applications in drug delivery because of their natural origin, biocompatibility, and ability to cross biological barriers. Using the unique properties of EVs could enhance drug accumulation at target sites, minimize systemic toxicity, and precisely target specific cells. Here, we discuss the characteristics and functionalization of EVs, the mechanisms of drug resistance, and the applications of engineered EVs to overcome drug resistance. This review provides a comprehensive overview of the advancements in EV-based drug delivery systems and their applications in overcoming cancer drug resistance. We highlight the potential of EV-based drug delivery systems to revolutionize cancer therapy and offer promising strategies for more effective treatment modalities.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"7 ","pages":"50"},"PeriodicalIF":4.6,"publicationDate":"2024-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11724354/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142973545","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Breast cancer cell resistance to hormonal and targeted therapeutics is correlated with the inactivation of the NR6A1 axis. 乳腺癌细胞对激素和靶向治疗的耐药性与NR6A1轴失活相关。
IF 4.6 Q1 ONCOLOGY Pub Date : 2024-11-23 eCollection Date: 2024-01-01 DOI: 10.20517/cdr.2024.69
Olga E Andreeva, Danila V Sorokin, Svetlana V Vinokurova, Pavel B Kopnin, Nadezhda V Elkina, Alexey N Katargin, Radik S Faskhutdinov, Diana I Salnikova, Alexander M Scherbakov, Mikhail A Krasil'nikov

Aim: Resistance to hormonal and targeted therapies in breast cancer limits treatment efficacy. Epigenetic alterations, including changes mediated by DNA methyltransferases, play a key role in this process. Previously, we identified that resistance to tamoxifen and rapamycin is associated with the suppression of DNMT3A. This study aims to further explore the mechanisms underlying this suppression, with a focus on identifying NR6A1 as a novel regulatory factor. Methods: Acquisition of resistant breast cancer cell sublines, MTT-test, immunoblotting, transient transfection and reporter analysis, lentiviral infection, qRT-PCR, and analysis of methylation using bisulfite pyrosequencing. Results: Our findings indicate that the development of cross-resistance in breast cancer cells to hormonal and targeted therapies involves a shift in cell signaling to alternative AKT pathways, marked by a localized suppression of the NR6A1/DNMT3A axis and associated DNA methylation changes. We demonstrated the critical role of NR6A1 downregulation in resistance development. Additionally, we observed activation of Snail - a key regulator in the epithelial-mesenchymal transition - as a mediator of the effects of NR6A1 depletion, establishing a direct link between Snail expression and resistance formation. Conclusion: The coordinated suppression of NR6A1 and DNMT3A may contribute to sustaining the resistant phenotype in breast cancer cells. This pathway could serve as a predictive marker, helping guide the selection of optimal therapeutic strategies for breast cancer treatment in the future.

目的:乳腺癌对激素和靶向治疗的耐药性限制了治疗效果。表观遗传改变,包括DNA甲基转移酶介导的变化,在这一过程中起关键作用。先前,我们发现对他莫昔芬和雷帕霉素的耐药性与DNMT3A的抑制有关。本研究旨在进一步探索这种抑制的机制,重点是确定NR6A1作为一种新的调节因子。方法:获得耐药乳腺癌细胞亚群,mtt试验,免疫印迹,瞬时转染和报告基因分析,慢病毒感染,qRT-PCR,亚硫酸氢盐焦磷酸测序分析甲基化。结果:我们的研究结果表明,乳腺癌细胞对激素和靶向治疗的交叉耐药的发展涉及细胞信号向替代AKT通路的转变,其标志是NR6A1/DNMT3A轴的局部抑制和相关的DNA甲基化变化。我们证明了NR6A1下调在耐药性发展中的关键作用。此外,我们观察到蜗牛的激活——上皮-间质转化的关键调节因子——作为NR6A1耗竭效应的中介,建立了蜗牛表达与抗性形成之间的直接联系。结论:NR6A1和DNMT3A的协同抑制可能有助于维持乳腺癌细胞的耐药表型。这一途径可以作为一种预测标志物,帮助指导未来乳腺癌治疗的最佳治疗策略的选择。
{"title":"Breast cancer cell resistance to hormonal and targeted therapeutics is correlated with the inactivation of the NR6A1 axis.","authors":"Olga E Andreeva, Danila V Sorokin, Svetlana V Vinokurova, Pavel B Kopnin, Nadezhda V Elkina, Alexey N Katargin, Radik S Faskhutdinov, Diana I Salnikova, Alexander M Scherbakov, Mikhail A Krasil'nikov","doi":"10.20517/cdr.2024.69","DOIUrl":"https://doi.org/10.20517/cdr.2024.69","url":null,"abstract":"<p><p><b>Aim:</b> Resistance to hormonal and targeted therapies in breast cancer limits treatment efficacy. Epigenetic alterations, including changes mediated by DNA methyltransferases, play a key role in this process. Previously, we identified that resistance to tamoxifen and rapamycin is associated with the suppression of DNMT3A. This study aims to further explore the mechanisms underlying this suppression, with a focus on identifying NR6A1 as a novel regulatory factor. <b>Methods:</b> Acquisition of resistant breast cancer cell sublines, MTT-test, immunoblotting, transient transfection and reporter analysis, lentiviral infection, qRT-PCR, and analysis of methylation using bisulfite pyrosequencing. <b>Results:</b> Our findings indicate that the development of cross-resistance in breast cancer cells to hormonal and targeted therapies involves a shift in cell signaling to alternative AKT pathways, marked by a localized suppression of the NR6A1/DNMT3A axis and associated DNA methylation changes. We demonstrated the critical role of NR6A1 downregulation in resistance development. Additionally, we observed activation of Snail - a key regulator in the epithelial-mesenchymal transition - as a mediator of the effects of NR6A1 depletion, establishing a direct link between Snail expression and resistance formation. <b>Conclusion:</b> The coordinated suppression of NR6A1 and DNMT3A may contribute to sustaining the resistant phenotype in breast cancer cells. This pathway could serve as a predictive marker, helping guide the selection of optimal therapeutic strategies for breast cancer treatment in the future.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"7 ","pages":"48"},"PeriodicalIF":4.6,"publicationDate":"2024-11-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11609143/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142775131","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mechanism of ferroptosis resistance in cancer cells. 癌细胞抗铁下垂机制的研究。
IF 4.6 Q1 ONCOLOGY Pub Date : 2024-11-20 eCollection Date: 2024-01-01 DOI: 10.20517/cdr.2024.127
Yuan Wang, Guifang Yu, Xin Chen

Ferroptosis is an iron-dependent cell death characterized by increased intracellular lipid peroxidation. Inducing ferroptosis has shown significant potential in eliminating various malignancies. However, the effectiveness of ferroptosis-based treatments is hampered by the intrinsic or acquired resistance of some tumors. In this review, we delineate the known mechanisms that regulate ferroptosis sensitivity and summarize the therapeutic application of ferroptosis inducers in cancer. Additionally, we discuss the roles of diverse signaling pathways that contribute to ferroptosis resistance in cancer cells, including the glutathione (GSH) and coenzyme Q (CoQ) pathways, NFE2-like bZIP transcription factor 2 (NRF2) antioxidant response, and lipid and iron metabolism. This emerging knowledge may serve as a foundation for developing novel anticancer strategies to overcome ferroptosis resistance.

铁下垂是一种铁依赖性细胞死亡,其特征是细胞内脂质过氧化增加。诱导铁下垂在消除各种恶性肿瘤方面显示出显著的潜力。然而,一些肿瘤的固有或获得性耐药阻碍了基于铁中毒的治疗的有效性。在这篇综述中,我们描述了已知的调节铁下垂敏感性的机制,并总结了铁下垂诱导剂在癌症中的治疗应用。此外,我们还讨论了多种信号通路的作用,包括谷胱甘肽(GSH)和辅酶Q (CoQ)途径,nfe2样bZIP转录因子2 (NRF2)抗氧化反应,以及脂质和铁代谢。这一新兴的知识可以作为开发新的抗癌策略来克服铁下垂抵抗的基础。
{"title":"Mechanism of ferroptosis resistance in cancer cells.","authors":"Yuan Wang, Guifang Yu, Xin Chen","doi":"10.20517/cdr.2024.127","DOIUrl":"https://doi.org/10.20517/cdr.2024.127","url":null,"abstract":"<p><p>Ferroptosis is an iron-dependent cell death characterized by increased intracellular lipid peroxidation. Inducing ferroptosis has shown significant potential in eliminating various malignancies. However, the effectiveness of ferroptosis-based treatments is hampered by the intrinsic or acquired resistance of some tumors. In this review, we delineate the known mechanisms that regulate ferroptosis sensitivity and summarize the therapeutic application of ferroptosis inducers in cancer. Additionally, we discuss the roles of diverse signaling pathways that contribute to ferroptosis resistance in cancer cells, including the glutathione (GSH) and coenzyme Q (CoQ) pathways, NFE2-like bZIP transcription factor 2 (NRF2) antioxidant response, and lipid and iron metabolism. This emerging knowledge may serve as a foundation for developing novel anticancer strategies to overcome ferroptosis resistance.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"7 ","pages":"47"},"PeriodicalIF":4.6,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11609146/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142775101","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
HDAC-driven mechanisms in anticancer resistance: epigenetics and beyond. hdac驱动的抗肿瘤机制:表观遗传学及其他。
IF 4.6 Q1 ONCOLOGY Pub Date : 2024-11-20 eCollection Date: 2024-01-01 DOI: 10.20517/cdr.2024.103
Martina Minisini, Martina Mascaro, Claudio Brancolini

The emergence of drug resistance leading to cancer recurrence is one of the challenges in the treatment of cancer patients. Several mechanisms can lead to drug resistance, including epigenetic changes. Histone deacetylases (HDACs) play a key role in chromatin regulation through epigenetic mechanisms and are also involved in drug resistance. The control of histone acetylation and the accessibility of regulatory DNA sequences such as promoters, enhancers, and super-enhancers are known mechanisms by which HDACs influence gene expression. Other targets of HDACs that are not histones can also contribute to resistance. This review describes the contribution of HDACs to the mechanisms that, in some cases, may determine resistance to chemotherapy or other cancer treatments.

耐药的出现导致癌症复发是癌症患者治疗面临的挑战之一。几种机制可导致耐药性,包括表观遗传变化。组蛋白去乙酰化酶(hdac)通过表观遗传机制在染色质调控中发挥关键作用,也参与耐药性。组蛋白乙酰化的控制和调控DNA序列(如启动子、增强子和超级增强子)的可及性是hdac影响基因表达的已知机制。hdac的其他非组蛋白靶点也可能导致耐药性。这篇综述描述了hdac在某些情况下可能决定化疗或其他癌症治疗耐药的机制中的作用。
{"title":"HDAC-driven mechanisms in anticancer resistance: epigenetics and beyond.","authors":"Martina Minisini, Martina Mascaro, Claudio Brancolini","doi":"10.20517/cdr.2024.103","DOIUrl":"https://doi.org/10.20517/cdr.2024.103","url":null,"abstract":"<p><p>The emergence of drug resistance leading to cancer recurrence is one of the challenges in the treatment of cancer patients. Several mechanisms can lead to drug resistance, including epigenetic changes. Histone deacetylases (HDACs) play a key role in chromatin regulation through epigenetic mechanisms and are also involved in drug resistance. The control of histone acetylation and the accessibility of regulatory DNA sequences such as promoters, enhancers, and super-enhancers are known mechanisms by which HDACs influence gene expression. Other targets of HDACs that are not histones can also contribute to resistance. This review describes the contribution of HDACs to the mechanisms that, in some cases, may determine resistance to chemotherapy or other cancer treatments.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"7 ","pages":"46"},"PeriodicalIF":4.6,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11609180/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142775097","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Reprogrammed lipid metabolism in advanced resistant cancers: an upcoming therapeutic opportunity. 重编程脂质代谢在晚期耐药癌症:一个即将到来的治疗机会。
IF 4.6 Q1 ONCOLOGY Pub Date : 2024-11-19 eCollection Date: 2024-01-01 DOI: 10.20517/cdr.2024.131
Mario Cioce, Mariamena Arbitrio, Nicoletta Polerà, Emanuela Altomare, Antonia Rizzuto, Carmela De Marco, Vito Michele Fazio, Giuseppe Viglietto, Maria Lucibello

Resistance of cancer to therapy is the main challenge to its therapeutic management and is still an unsolved problem. Rearranged lipid metabolism is a strategy adopted by cancer cells to counteract adversity during their evolution toward aggressiveness and immune evasion. This relies on several mechanisms, ranging from altered metabolic pathways within cancer cells to evolved dynamic crosstalk between cancer cells and the tumor microenvironment (TME), with some cell populations at the forefront of metabolic reprogramming, thereby contributing to the resistance of the whole ecosystem during therapy. Unraveling these mechanisms may contribute to the development of more effective combinatorial therapy in resistant patients. This review highlights the alterations in lipid metabolism that contribute to cancer progression, with a focus on the potential clinical relevance of such findings for the management of therapy resistance.

肿瘤对治疗的抵抗是对其治疗管理的主要挑战,也是一个尚未解决的问题。脂质代谢的重排是癌细胞在向侵袭性和免疫逃避进化过程中对抗逆境的一种策略。这依赖于几种机制,从癌细胞内代谢途径的改变到癌细胞与肿瘤微环境(TME)之间进化的动态串扰,一些细胞群处于代谢重编程的前沿,从而在治疗期间促进整个生态系统的抵抗。解开这些机制可能有助于开发更有效的联合治疗耐药患者。这篇综述强调了导致癌症进展的脂质代谢的改变,重点是这些发现与治疗耐药管理的潜在临床相关性。
{"title":"Reprogrammed lipid metabolism in advanced resistant cancers: an upcoming therapeutic opportunity.","authors":"Mario Cioce, Mariamena Arbitrio, Nicoletta Polerà, Emanuela Altomare, Antonia Rizzuto, Carmela De Marco, Vito Michele Fazio, Giuseppe Viglietto, Maria Lucibello","doi":"10.20517/cdr.2024.131","DOIUrl":"https://doi.org/10.20517/cdr.2024.131","url":null,"abstract":"<p><p>Resistance of cancer to therapy is the main challenge to its therapeutic management and is still an unsolved problem. Rearranged lipid metabolism is a strategy adopted by cancer cells to counteract adversity during their evolution toward aggressiveness and immune evasion. This relies on several mechanisms, ranging from altered metabolic pathways within cancer cells to evolved dynamic crosstalk between cancer cells and the tumor microenvironment (TME), with some cell populations at the forefront of metabolic reprogramming, thereby contributing to the resistance of the whole ecosystem during therapy. Unraveling these mechanisms may contribute to the development of more effective combinatorial therapy in resistant patients. This review highlights the alterations in lipid metabolism that contribute to cancer progression, with a focus on the potential clinical relevance of such findings for the management of therapy resistance.</p>","PeriodicalId":70759,"journal":{"name":"癌症耐药(英文)","volume":"7 ","pages":"45"},"PeriodicalIF":4.6,"publicationDate":"2024-11-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11609178/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142775104","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
癌症耐药(英文)
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