首页 > 最新文献

Recent patents on anti-cancer drug discovery最新文献

英文 中文
The m6A Reader IGF2BP2 Promotes Oral Squamous Cell Carcinoma Progression by Maintaining UCA1 Stability. m6A 读取器 IGF2BP2 通过维持 UCA1 的稳定性促进口腔鳞状细胞癌的发展。
Pub Date : 2024-08-26 DOI: 10.2174/0115748928293003240817180839
Duheyi Zhang, Lianxi Mai, Lizao Zhang, Guoxin Huang, Zhaoyu Lin, Shuang Wang, Guangxin Rao, Shule Xie, Chaobin Pan

Introduction: N6-methyladenosine (m6A) modifications of RNAs are associated with many cancer types. Nevertheless, the function of the m6A reader IGF2BP2 in oral squamous cell carcinoma (OSCC) has yet to be ascertained.

Aims: The objective of this investigation was to elucidate the role of IGF2BP2 in OSCC and delineate the associated mechanisms.

Method: Elevated expression of IGF2BP2 was observed in OSCC, and this overexpression significantly correlated with adverse prognostic outcomes in patients with OSCC. In vitro analyses demonstrated that silencing of IGF2BP2 attenuated the proliferation, migration, and invasion capabilities of oral cancer cells while concurrently promoting apoptosis.

Results: In vivo experiments demonstrated that IGF2BP2 promoted OSCC growth. RNA-seq and m6A-seq were utilized to elucidate the downstream targets of IGF2BP2. Through bioinformatic analysis, we identified the long noncoding RNA (lncRNA) UCA1 as a target. IGF2BP2 was found to maintain the stability of UCA1 in an m6A-dependent manner by binding to m6A-modified UCA1 and plays an oncogenic role in OSCC through UCA1.

Conclusion: In conclusion, we identified IGF2BP2 as a prognostic biomarker of OSCC, and the IGF2BP2-UCA1 axis was found to promote OSCC progression and may perform as a novel therapeutic target.

简介RNA的N6-甲基腺苷(m6A)修饰与许多癌症类型有关。然而,m6A阅读器IGF2BP2在口腔鳞状细胞癌(OSCC)中的功能尚待确定:方法:在OSCC中观察到IGF2BP2的表达升高,这种过表达与OSCC患者的不良预后结果显著相关。体外分析表明,沉默 IGF2BP2 可减轻口腔癌细胞的增殖、迁移和侵袭能力,同时促进细胞凋亡:体内实验表明,IGF2BP2 促进了 OSCC 的生长。我们利用 RNA-seq 和 m6A-seq 阐明了 IGF2BP2 的下游靶标。通过生物信息学分析,我们发现长非编码 RNA(lncRNA)UCA1 是一个靶点。研究发现,IGF2BP2通过与m6A修饰的UCA1结合,以m6A依赖性方式维持UCA1的稳定性,并通过UCA1在OSCC中发挥致癌作用:总之,我们发现IGF2BP2是OSCC的预后生物标志物,IGF2BP2-UCA1轴促进了OSCC的进展,可作为新的治疗靶点。
{"title":"The m6A Reader IGF2BP2 Promotes Oral Squamous Cell Carcinoma Progression by Maintaining UCA1 Stability.","authors":"Duheyi Zhang, Lianxi Mai, Lizao Zhang, Guoxin Huang, Zhaoyu Lin, Shuang Wang, Guangxin Rao, Shule Xie, Chaobin Pan","doi":"10.2174/0115748928293003240817180839","DOIUrl":"https://doi.org/10.2174/0115748928293003240817180839","url":null,"abstract":"<p><strong>Introduction: </strong>N6-methyladenosine (m6A) modifications of RNAs are associated with many cancer types. Nevertheless, the function of the m6A reader IGF2BP2 in oral squamous cell carcinoma (OSCC) has yet to be ascertained.</p><p><strong>Aims: </strong>The objective of this investigation was to elucidate the role of IGF2BP2 in OSCC and delineate the associated mechanisms.</p><p><strong>Method: </strong>Elevated expression of IGF2BP2 was observed in OSCC, and this overexpression significantly correlated with adverse prognostic outcomes in patients with OSCC. In vitro analyses demonstrated that silencing of IGF2BP2 attenuated the proliferation, migration, and invasion capabilities of oral cancer cells while concurrently promoting apoptosis.</p><p><strong>Results: </strong>In vivo experiments demonstrated that IGF2BP2 promoted OSCC growth. RNA-seq and m6A-seq were utilized to elucidate the downstream targets of IGF2BP2. Through bioinformatic analysis, we identified the long noncoding RNA (lncRNA) UCA1 as a target. IGF2BP2 was found to maintain the stability of UCA1 in an m6A-dependent manner by binding to m6A-modified UCA1 and plays an oncogenic role in OSCC through UCA1.</p><p><strong>Conclusion: </strong>In conclusion, we identified IGF2BP2 as a prognostic biomarker of OSCC, and the IGF2BP2-UCA1 axis was found to promote OSCC progression and may perform as a novel therapeutic target.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142082941","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Comprehensive Analysis and Experimental Validation of TLL2 as a Potential New Prognostic Biomarker Associated with Immune Infiltration in Lung Adenocarcinoma. 将 TLL2 作为与肺腺癌免疫浸润相关的潜在新预后生物标记物的综合分析和实验验证
Pub Date : 2024-08-26 DOI: 10.2174/0115748928303392240817131807
Jing Jiang, Zhongye Du, Haijuan Tang, Yingying Huang, Dongbing Li, Qiuli Liang

Background: The precise function of Tolloid Like 2 (TLL2) remains uncertain within the context of Lung Adenocarcinoma (LUAD).

Objective: The primary objective of this investigation was to conduct a thorough analysis.

Methods: To assess its diagnostic utility, data from The Cancer Genome Atlas (TCGA) database were used to assess TLL2 expression in pan-cancer and LUAD. The study has also investigated the correlation between TLL2 expression levels and LUAD symptoms and prognosis. Furthermore, the study has explored possible regulatory networks involving TLL2, including its association with immune infiltration, tumor stemness index (mRNAsi), and drug sensitivity in LUAD. We have explored TLL2 expression in single-cell sequencing of LUAD and the genomic variation and clinical significance of TLL2 in LUAD. The expression of TLL2 has been validated in GSE87340 and cell lines by quantitative Real-time PCR (qRT-PCR).

Results: An abnormal expression of TLL2 has been found in pan-cancer and LUAD. In LUAD patients, elevated levels of TLL2 were significantly related to the T stage (p = 0.046) and the pathological stage (p = 0.016). The expression of TLL2 in patients with LUAD was significantly associated with poorer Overall Survival (OS) (p < 0.001). The expression of TLL2 was determined to be an independent predictor of poorer OS (p = 0.042). TLL2 was associated with ribosome, neuroactive ligand-receptor interaction, allograft rejection, ECM receptor interaction, asthma, porphyrin and chlorophyll metabolism, focal adhesion, pentose and glucuronate inter-conversions, and ascorbate and aldarate metabolism. The expression of TLL2 in LUAD was correlated with immune infiltration and mRNAsi. The expression of TLL2 was significantly and negatively correlated with TAK-715, XMD13-2, STF-62247, OSI-930, and EHT-1864 in LUAD. The TLL2 gene was up- -regulated in multiple individual LUAD cells. LUAD patients with altered TLL2 had a shorter PFS as opposed to those with unaltered TLL2. The expression of TLL2 was significantly increased in LUAD cells.

Conclusion: For patients with LUAD, TLL2 may serve as an immunotherapeutic target and a useful prognosis biomarker.

背景:在肺腺癌(LUAD)的背景下,类甲状腺球蛋白2(TLL2)的确切功能仍不确定:本研究的主要目的是进行全面分析:为了评估其诊断效用,研究人员使用了癌症基因组图谱(TCGA)数据库中的数据来评估TLL2在泛癌症和LUAD中的表达。研究还调查了 TLL2 表达水平与 LUAD 症状和预后之间的相关性。此外,研究还探讨了涉及 TLL2 的可能调控网络,包括其与免疫浸润、肿瘤干性指数(mRNAsi)和 LUAD 药物敏感性的关联。我们探讨了 TLL2 在 LUAD 单细胞测序中的表达,以及 TLL2 在 LUAD 中的基因组变异和临床意义。通过实时定量 PCR(qRT-PCR)技术验证了 TLL2 在 GSE87340 和细胞系中的表达:结果:在泛癌和 LUAD 中发现了 TLL2 的异常表达。在 LUAD 患者中,TLL2 水平的升高与 T 分期(p = 0.046)和病理分期(p = 0.016)显著相关。TLL2在LUAD患者中的表达与较差的总生存期(OS)明显相关(p < 0.001)。TLL2的表达被确定为较差OS的独立预测因子(p = 0.042)。TLL2与核糖体、神经活性配体-受体相互作用、异体移植排斥反应、ECM受体相互作用、哮喘、卟啉和叶绿素代谢、病灶粘附、戊糖和葡萄糖醛酸相互转化以及抗坏血酸和醛酸代谢有关。TLL2 在 LUAD 中的表达与免疫浸润和 mRNAsi 相关。在 LUAD 中,TLL2 的表达与 TAK-715、XMD13-2、STF-62247、OSI-930 和 EHT-1864 呈显著负相关。在多个 LUAD 细胞中,TLL2 基因被上调。与TLL2基因未发生改变的LUAD患者相比,TLL2基因发生改变的LUAD患者的PFS较短。TLL2在LUAD细胞中的表达明显增加:结论:对于 LUAD 患者,TLL2 可作为免疫治疗靶点和有用的预后生物标志物。
{"title":"Comprehensive Analysis and Experimental Validation of TLL2 as a Potential New Prognostic Biomarker Associated with Immune Infiltration in Lung Adenocarcinoma.","authors":"Jing Jiang, Zhongye Du, Haijuan Tang, Yingying Huang, Dongbing Li, Qiuli Liang","doi":"10.2174/0115748928303392240817131807","DOIUrl":"https://doi.org/10.2174/0115748928303392240817131807","url":null,"abstract":"<p><strong>Background: </strong>The precise function of Tolloid Like 2 (TLL2) remains uncertain within the context of Lung Adenocarcinoma (LUAD).</p><p><strong>Objective: </strong>The primary objective of this investigation was to conduct a thorough analysis.</p><p><strong>Methods: </strong>To assess its diagnostic utility, data from The Cancer Genome Atlas (TCGA) database were used to assess TLL2 expression in pan-cancer and LUAD. The study has also investigated the correlation between TLL2 expression levels and LUAD symptoms and prognosis. Furthermore, the study has explored possible regulatory networks involving TLL2, including its association with immune infiltration, tumor stemness index (mRNAsi), and drug sensitivity in LUAD. We have explored TLL2 expression in single-cell sequencing of LUAD and the genomic variation and clinical significance of TLL2 in LUAD. The expression of TLL2 has been validated in GSE87340 and cell lines by quantitative Real-time PCR (qRT-PCR).</p><p><strong>Results: </strong>An abnormal expression of TLL2 has been found in pan-cancer and LUAD. In LUAD patients, elevated levels of TLL2 were significantly related to the T stage (p = 0.046) and the pathological stage (p = 0.016). The expression of TLL2 in patients with LUAD was significantly associated with poorer Overall Survival (OS) (p < 0.001). The expression of TLL2 was determined to be an independent predictor of poorer OS (p = 0.042). TLL2 was associated with ribosome, neuroactive ligand-receptor interaction, allograft rejection, ECM receptor interaction, asthma, porphyrin and chlorophyll metabolism, focal adhesion, pentose and glucuronate inter-conversions, and ascorbate and aldarate metabolism. The expression of TLL2 in LUAD was correlated with immune infiltration and mRNAsi. The expression of TLL2 was significantly and negatively correlated with TAK-715, XMD13-2, STF-62247, OSI-930, and EHT-1864 in LUAD. The TLL2 gene was up- -regulated in multiple individual LUAD cells. LUAD patients with altered TLL2 had a shorter PFS as opposed to those with unaltered TLL2. The expression of TLL2 was significantly increased in LUAD cells.</p><p><strong>Conclusion: </strong>For patients with LUAD, TLL2 may serve as an immunotherapeutic target and a useful prognosis biomarker.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142082993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Phosphofructokinase-1 in Cancer: A Promising Target for Diagnosis and Therapy. 癌症中的磷酸果糖激酶-1:有望成为诊断和治疗的靶点
Pub Date : 2024-08-21 DOI: 10.2174/0115748928321372240813080131
Ali Raza Ishaq, Tahira Younis, Shankun Lin, Muhammad Usman, Tingfang Wang, Zhe-Sheng Chen

Tumor cells have distorted enzymatic houses, which change the metabolic state from oxidative phosphorylation to glycolysis with high lactate levels in a hypoxic environment. Redrafting the metabolic profile is an emerging hallmark of cancer. Glycolytic enzyme amplification occurs in about 70% of all malignancies. Current studies have found that PFK-1 overexpression is linked to cell migration, proliferation, and Overall Survival (OS) rate in various human cancer cell lines. This review intended to uncover the bona fide therapeutic target for cancer therapy and elucidate the role of PFK-1 in cancer. Furthermore, this review has outlined the listed pharmacological and genetic inhibitors of PFK-1. Following this review, future studies on PFK-1 should emphasize the molecular pathways implicated in PFK-1 overexpression in cancer development. The terms "PFK-1", "PFKP-1", "PFKL-1", "PFKM-1", "PFKM-1 and cancer", "PFKP-1 and cancer", "PFKL-1 and cancer", and "inhibitors of PFK-1" were used to retrieve the information from a variety of databases, including PubMed, Scopus, Google Scholar, and ScienceDirect. In a variety of malignancies, inhibiting the expression of PFK-1 isoforms has been reported to be the most effective therapeutic method. Overexpression of PFK-1 isoforms induces the Warburg effect, cell proliferation, and carcinogenesis by downregulating apoptotic proteins, such as active caspase-3, caspase-9, and caspase-8. YY1, synoviolin, Sh-RNA-507, SNAI, miR-520a/b/e, miR-128, and β-miR-6517 are some of the putative genetic inhibitors against PFK-1 that have been used to manage the development of malignancies. Pharmacological inhibitors, such as penfluridol, synoviolin/HRD1, quercetin, ginsenoside 20(S)-Rg3, triptolide, worenine, acetylsalicylic acid, and salicylic acid, can regulate the advancement of malignancies by inhibiting PFK-1. Thus, PFK-1 is a promising molecular biomarker for cancer treatment. A prospective investigation can validate the unbiased approaches for discovering brandnew PFK-1 inhibitors for cancer treatment.

在缺氧环境中,肿瘤细胞的酶系统发生扭曲,代谢状态从氧化磷酸化转变为高乳酸水平的糖酵解。新陈代谢谱的重写是癌症的一个新特征。约 70% 的恶性肿瘤都会出现糖酵解酶扩增。目前的研究发现,在各种人类癌细胞系中,PFK-1 的过表达与细胞迁移、增殖和总生存率(OS)有关。本综述旨在揭示癌症治疗的真正治疗靶点,并阐明 PFK-1 在癌症中的作用。此外,本综述还列出了 PFK-1 的药理和基因抑制剂。在本综述之后,未来有关 PFK-1 的研究应强调癌症发展过程中 PFK-1 过度表达所涉及的分子通路。我们使用了 "PFK-1"、"PFKP-1"、"PFKL-1"、"PFKM-1"、"PFKM-1 与癌症"、"PFKP-1 与癌症"、"PFKL-1 与癌症 "和 "PFK-1 抑制剂 "等术语,从 PubMed、Scopus、Google Scholar 和 ScienceDirect 等多个数据库中检索信息。据报道,在多种恶性肿瘤中,抑制 PFK-1 同工酶的表达是最有效的治疗方法。PFK-1 同工酶的过度表达会诱导沃伯格效应、细胞增殖,并通过下调活性 caspase-3、caspase-9 和 caspase-8 等凋亡蛋白而致癌。YY1、synoviolin、Sh-RNA-507、SNAI、miR-520a/b/e、miR-128 和 β-miR-6517 是一些针对 PFK-1 的假定基因抑制剂,已被用于控制恶性肿瘤的发展。药理抑制剂,如五氟利多、滑石素/HRD1、槲皮素、人参皂苷 20(S)-Rg3、三七皂苷、沃仑宁、乙酰水杨酸和水杨酸,可通过抑制 PFK-1 来调节恶性肿瘤的发展。因此,PFK-1 是一种很有前景的癌症治疗分子生物标记物。前瞻性研究可以验证发现治疗癌症的全新 PFK-1 抑制剂的无偏见方法。
{"title":"Phosphofructokinase-1 in Cancer: A Promising Target for Diagnosis and Therapy.","authors":"Ali Raza Ishaq, Tahira Younis, Shankun Lin, Muhammad Usman, Tingfang Wang, Zhe-Sheng Chen","doi":"10.2174/0115748928321372240813080131","DOIUrl":"https://doi.org/10.2174/0115748928321372240813080131","url":null,"abstract":"<p><p>Tumor cells have distorted enzymatic houses, which change the metabolic state from oxidative phosphorylation to glycolysis with high lactate levels in a hypoxic environment. Redrafting the metabolic profile is an emerging hallmark of cancer. Glycolytic enzyme amplification occurs in about 70% of all malignancies. Current studies have found that PFK-1 overexpression is linked to cell migration, proliferation, and Overall Survival (OS) rate in various human cancer cell lines. This review intended to uncover the bona fide therapeutic target for cancer therapy and elucidate the role of PFK-1 in cancer. Furthermore, this review has outlined the listed pharmacological and genetic inhibitors of PFK-1. Following this review, future studies on PFK-1 should emphasize the molecular pathways implicated in PFK-1 overexpression in cancer development. The terms \"PFK-1\", \"PFKP-1\", \"PFKL-1\", \"PFKM-1\", \"PFKM-1 and cancer\", \"PFKP-1 and cancer\", \"PFKL-1 and cancer\", and \"inhibitors of PFK-1\" were used to retrieve the information from a variety of databases, including PubMed, Scopus, Google Scholar, and ScienceDirect. In a variety of malignancies, inhibiting the expression of PFK-1 isoforms has been reported to be the most effective therapeutic method. Overexpression of PFK-1 isoforms induces the Warburg effect, cell proliferation, and carcinogenesis by downregulating apoptotic proteins, such as active caspase-3, caspase-9, and caspase-8. YY1, synoviolin, Sh-RNA-507, SNAI, miR-520a/b/e, miR-128, and β-miR-6517 are some of the putative genetic inhibitors against PFK-1 that have been used to manage the development of malignancies. Pharmacological inhibitors, such as penfluridol, synoviolin/HRD1, quercetin, ginsenoside 20(S)-Rg3, triptolide, worenine, acetylsalicylic acid, and salicylic acid, can regulate the advancement of malignancies by inhibiting PFK-1. Thus, PFK-1 is a promising molecular biomarker for cancer treatment. A prospective investigation can validate the unbiased approaches for discovering brandnew PFK-1 inhibitors for cancer treatment.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142019986","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ligustilide Inhibits the PI3K/AKT Signalling Pathway and Suppresses Cholangiocarcinoma Cell Proliferation, Migration, and Invasion. 利格列汀抑制 PI3K/AKT 信号通路并抑制胆管癌细胞增殖、迁移和侵袭
Pub Date : 2024-08-20 DOI: 10.2174/0115748928332384240812060751
Yue Wu, Li Rong, Suifeng Zhang, Yuxi He, Na Song, Guoqing Zuo, Zhechuan Mei

Background: Angelica sinensis (Oliv.) Diels, a renowned traditional Chinese medicine, has gained widespread recognition for its antitumor properties. Further investigation is warranted to determine whether ligustilide (LIG), which is extracted from this plant, can effectively inhibit tumors.

Objective: We delved into the impact of LIG on cholangiocarcinoma cells, aiming to unravel the mechanisms underlying its effects.

Materials and methods: Cholangiocarcinoma cells (HuccT1 and RBE) were exposed to varying concentrations of LIG (2, 5, 10, 15, 20 μg/mL) for 24, 48, and 72 h. After identifying differentially expressed genes, stable transcription strains were utilized to explore LIG's antitumor mechanism. The inhibitory effects of LIG (5 μg/mL, 48 h) were assessed by CCK-8, colony formation, wound healing, transwell migration, western blotting, and immunofluorescence. In vivo, experiments in NOG mice (Ac, Ac+LIG; five per group) evaluated LIG's antiproliferative efficacy (5 mg/kg, intraperitoneal injection, 18-day period).

Results: LIG significantly inhibited cell proliferation and migration with IC50 5.08 and 5.77 μg/mL in HuccT1 and RBE cell lines at 48h, increased the expression of E-cadherin while decreased N-cadherin and the protein of PI3K/AKT pathway. Silenced NDRG1 (N-Myc downstream- regulated gene 1) attenuated these effects. In vivo, the AC+LIG group (LIG, 5 mg/kg, qd, 18 d) exhibited smaller tumor volumes compared to the Ac group. The expression of Ki-67 was significantly downregulated in the AC+LIG group.

Conclusion: For the first time, our study has revealed that LIG holds therapeutic potential for treating cholangiocarcinoma. These findings hold promise for advancing innovative therapeutic approaches in the treatment of cholangiocarcinoma. LIG may serve as a useful patent for treating CCA.

背景:当归(Oliv. Diels)是一种著名的传统中药,其抗肿瘤特性已得到广泛认可。从这种植物中提取的藁本内酯(LIG)是否能有效抑制肿瘤,还有待进一步研究:我们深入研究了藁本内酯对胆管癌细胞的影响,旨在揭示其作用机制:将胆管癌细胞(HuccT1和RBE)暴露于不同浓度的LIG(2、5、10、15、20 μg/mL)中24、48和72小时。LIG(5 μg/mL,48 小时)的抑制作用通过 CCK-8、菌落形成、伤口愈合、Transwell 迁移、Western 印迹和免疫荧光进行评估。在 NOG 小鼠(Ac、Ac+LIG;每组 5 只)体内进行的实验评估了 LIG 的抗增殖功效(5 毫克/千克,腹腔注射,18 天):48小时后,LIG能明显抑制HuccT1和RBE细胞株的增殖和迁移,IC50分别为5.08和5.77 μg/mL,并能增加E-cadherin的表达,同时降低N-cadherin和PI3K/AKT通路蛋白的表达。抑制 NDRG1(N-Myc 下游调控基因 1)可减轻这些影响。在体内,AC+LIG 组(LIG,5 mg/kg,qd,18 d)的肿瘤体积比 Ac 组小。AC+LIG组的Ki-67表达明显下调:我们的研究首次揭示了 LIG 治疗胆管癌的潜力。这些发现有望推动胆管癌治疗方法的创新。LIG 可作为治疗 CCA 的有效专利。
{"title":"Ligustilide Inhibits the PI3K/AKT Signalling Pathway and Suppresses Cholangiocarcinoma Cell Proliferation, Migration, and Invasion.","authors":"Yue Wu, Li Rong, Suifeng Zhang, Yuxi He, Na Song, Guoqing Zuo, Zhechuan Mei","doi":"10.2174/0115748928332384240812060751","DOIUrl":"https://doi.org/10.2174/0115748928332384240812060751","url":null,"abstract":"<p><strong>Background: </strong>Angelica sinensis (Oliv.) Diels, a renowned traditional Chinese medicine, has gained widespread recognition for its antitumor properties. Further investigation is warranted to determine whether ligustilide (LIG), which is extracted from this plant, can effectively inhibit tumors.</p><p><strong>Objective: </strong>We delved into the impact of LIG on cholangiocarcinoma cells, aiming to unravel the mechanisms underlying its effects.</p><p><strong>Materials and methods: </strong>Cholangiocarcinoma cells (HuccT1 and RBE) were exposed to varying concentrations of LIG (2, 5, 10, 15, 20 μg/mL) for 24, 48, and 72 h. After identifying differentially expressed genes, stable transcription strains were utilized to explore LIG's antitumor mechanism. The inhibitory effects of LIG (5 μg/mL, 48 h) were assessed by CCK-8, colony formation, wound healing, transwell migration, western blotting, and immunofluorescence. In vivo, experiments in NOG mice (Ac, Ac+LIG; five per group) evaluated LIG's antiproliferative efficacy (5 mg/kg, intraperitoneal injection, 18-day period).</p><p><strong>Results: </strong>LIG significantly inhibited cell proliferation and migration with IC50 5.08 and 5.77 μg/mL in HuccT1 and RBE cell lines at 48h, increased the expression of E-cadherin while decreased N-cadherin and the protein of PI3K/AKT pathway. Silenced NDRG1 (N-Myc downstream- regulated gene 1) attenuated these effects. In vivo, the AC+LIG group (LIG, 5 mg/kg, qd, 18 d) exhibited smaller tumor volumes compared to the Ac group. The expression of Ki-67 was significantly downregulated in the AC+LIG group.</p><p><strong>Conclusion: </strong>For the first time, our study has revealed that LIG holds therapeutic potential for treating cholangiocarcinoma. These findings hold promise for advancing innovative therapeutic approaches in the treatment of cholangiocarcinoma. LIG may serve as a useful patent for treating CCA.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142019985","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ruthenium Complex Suppresses Proliferation of Residual Hepatocellular Carcinoma after Incomplete Radiofrequency Ablation Therapy. 钌复合物能抑制不完全射频消融治疗后残留肝细胞癌的增殖
Pub Date : 2024-08-12 DOI: 10.2174/0115748928320508240802055846
Zhi-Jie Yu, Shun-Wen Guo, Bi-Shu Wang, Shi Ouyang, Xian-Huan Zhang, Zi-Zhuo Zhao, Jin-Quan Wang

Background: Radiofrequency ablation (RFA) is an effective therapy for hepatocellular carcinoma (HCC). However, incomplete radiofrequency ablation (IRFA) can promote the progression of residual cancer cells, which is a serious problem in the clinical application of RFA. Therefore, it is of great significance to explore the mechanism and countermeasures of the progression of residual tumors after IRFA. Our previous study confirmed that IRFA can activate the hypoxia/ autophagy pathway of residual tumors in mice and then induce the proliferation of residual tumor cells. Additionally, we found a metal ruthenium complex [Ru(bpy)2(ipad)](ClO4)2 (Ru, where bpy = 2,2'-bipyridine and ipad = 2-(anthracene-9,10-dione-2-yl)imidazo[4,5-f][1,10]phenanthroline) can effectively inhibit hypoxia-inducible factor (HIF-1α) and has good anti-tumor effect in a hypoxic environment; however, whether Ru could suppress the proliferation of residual tumor cells after IRFA is unknown.

Objective: This study intends to evaluate the effect of Ru in suppressing the proliferation of residual hepatocellular carcinoma after IRFA in a mice model.

Methods: The Hepa1-6 xenograft mouse model was established in C57BL/6 mice to simulate clinical IRFA. H&E staining was used to evaluate the biosafety of major organs in the treated mice. TUNEL assay was employed to assess the antitumor effect. Immunohistochemically and immunofluorescence staining was performed to detect the expression of HIF-1α and autophagy-related proteins. The ELISA assay was used to examine the cytokines of interferon-gamma (IFN-γ) and interleukin 10 (IL-10).

Results: Our findings revealed that the residual tumor relapsed via the HIF-1α/LC3B/P62 autophagy- related pathway after IRFA, while Ru could suppress this process. In addition, it was demonstrated that Ru could effectively activate the immune system of the mice and reverse the tumor immune suppression microenvironment after IRFA.

Conclusion: The ruthenium complex Ru could suppress the proliferation of residual hepatocellular carcinoma cells after IRFA in the mice model. This study introduces a novel approach that combines the use of ruthenium complexes with IRFA, offering a potential solution to address the reoccurrence of residual liver cancer following IRFA in clinical settings.

背景:射频消融(RFA)是治疗肝细胞癌(HCC)的有效方法。然而,不完全射频消融(IRFA)会促进残留癌细胞的进展,这是 RFA 临床应用中的一个严重问题。因此,探讨 IRFA 后残留肿瘤进展的机制和对策具有重要意义。我们之前的研究证实,IRFA能激活小鼠残留肿瘤的缺氧/自噬通路,进而诱导残留肿瘤细胞的增殖。此外,我们还发现一种金属钌复合物[Ru(bpy)2(ipad)](ClO4)2(Ru,其中 bpy = 2,2'-联吡啶,ipad = 2-(蒽-9,10-二酮-2-基)咪唑并[4,5-f][1,10]菲罗啉)能有效抑制缺氧诱导因子(HIF-1α),在缺氧环境中具有良好的抗肿瘤作用;然而,Ru 能否抑制 IRFA 后残余肿瘤细胞的增殖尚不清楚。研究目的本研究旨在评估 Ru 在小鼠模型中抑制 IRFA 后残余肝癌细胞增殖的效果:方法:在 C57BL/6 小鼠中建立 Hepa1-6 异种移植小鼠模型,以模拟临床 IRFA。采用 H&E 染色法评估治疗小鼠主要器官的生物安全性。TUNEL检测用于评估抗肿瘤效果。免疫组化和免疫荧光染色检测 HIF-1α 和自噬相关蛋白的表达。用 ELISA 法检测细胞因子γ干扰素(IFN-γ)和白细胞介素 10(IL-10):结果:我们的研究结果表明,IRFA 后残余肿瘤通过 HIF-1α/LC3B/P62 自噬相关途径复发,而 Ru 能抑制这一过程。此外,研究还表明,Ru能有效激活小鼠的免疫系统,逆转IRFA后的肿瘤免疫抑制微环境:结论:钌复合物 Ru 可抑制小鼠模型 IRFA 后残余肝癌细胞的增殖。本研究介绍了一种将钌复合物与 IRFA 结合使用的新方法,为临床上解决 IRFA 后残余肝癌再发问题提供了一种潜在的解决方案。
{"title":"Ruthenium Complex Suppresses Proliferation of Residual Hepatocellular Carcinoma after Incomplete Radiofrequency Ablation Therapy.","authors":"Zhi-Jie Yu, Shun-Wen Guo, Bi-Shu Wang, Shi Ouyang, Xian-Huan Zhang, Zi-Zhuo Zhao, Jin-Quan Wang","doi":"10.2174/0115748928320508240802055846","DOIUrl":"https://doi.org/10.2174/0115748928320508240802055846","url":null,"abstract":"<p><strong>Background: </strong>Radiofrequency ablation (RFA) is an effective therapy for hepatocellular carcinoma (HCC). However, incomplete radiofrequency ablation (IRFA) can promote the progression of residual cancer cells, which is a serious problem in the clinical application of RFA. Therefore, it is of great significance to explore the mechanism and countermeasures of the progression of residual tumors after IRFA. Our previous study confirmed that IRFA can activate the hypoxia/ autophagy pathway of residual tumors in mice and then induce the proliferation of residual tumor cells. Additionally, we found a metal ruthenium complex [Ru(bpy)2(ipad)](ClO4)2 (Ru, where bpy = 2,2'-bipyridine and ipad = 2-(anthracene-9,10-dione-2-yl)imidazo[4,5-f][1,10]phenanthroline) can effectively inhibit hypoxia-inducible factor (HIF-1α) and has good anti-tumor effect in a hypoxic environment; however, whether Ru could suppress the proliferation of residual tumor cells after IRFA is unknown.</p><p><strong>Objective: </strong>This study intends to evaluate the effect of Ru in suppressing the proliferation of residual hepatocellular carcinoma after IRFA in a mice model.</p><p><strong>Methods: </strong>The Hepa1-6 xenograft mouse model was established in C57BL/6 mice to simulate clinical IRFA. H&E staining was used to evaluate the biosafety of major organs in the treated mice. TUNEL assay was employed to assess the antitumor effect. Immunohistochemically and immunofluorescence staining was performed to detect the expression of HIF-1α and autophagy-related proteins. The ELISA assay was used to examine the cytokines of interferon-gamma (IFN-γ) and interleukin 10 (IL-10).</p><p><strong>Results: </strong>Our findings revealed that the residual tumor relapsed via the HIF-1α/LC3B/P62 autophagy- related pathway after IRFA, while Ru could suppress this process. In addition, it was demonstrated that Ru could effectively activate the immune system of the mice and reverse the tumor immune suppression microenvironment after IRFA.</p><p><strong>Conclusion: </strong>The ruthenium complex Ru could suppress the proliferation of residual hepatocellular carcinoma cells after IRFA in the mice model. This study introduces a novel approach that combines the use of ruthenium complexes with IRFA, offering a potential solution to address the reoccurrence of residual liver cancer following IRFA in clinical settings.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141972451","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identification of Immune Infiltration-related Molecular Features in Ovarian Cancer Patients and Experimental Validation of Immune Response Molecular Mechanisms through Integrated WGCNA, Machine Learning, and Single-cell Sequencing Analysis. 通过整合 WGCNA、机器学习和单细胞测序分析,识别卵巢癌患者免疫浸润相关分子特征并对免疫反应分子机制进行实验验证
Pub Date : 2024-07-18 DOI: 10.2174/0115748928297769240611055258
Juan Yang, Chengli Wen, Ping Li, Mingxiao Yao, Jing Wang

Background: Ovarian cancer is one of the most common gynecological malignancies globally, and immunotherapy has emerged as a promising treatment strategy in recent years. However, the effectiveness of immunotherapy is often limited by immune escape mechanisms.

Objective: To unravel the immune response mechanisms in ovarian cancer, this study aimed to employ integrated Weighted Gene Co-expression Network Analysis (WGCNA), machine learning, and single-- cell sequencing analysis to systematically investigate immune infiltration-related molecular features in ovarian cancer patients and experimentally validate the molecular mechanisms of the immune response. This research may provide a new theoretical foundation and treatment strategy for immune-based therapies in ovarian cancer.

Methods: Relevant ovarian cancer datasets were collected from public databases. The ConsensusCluster- Plus and ggplot2 R packages were used to perform dimensionality reduction and clustering analysis of immune infiltration-related genes. Various algorithms were employed to select the best ovarian cancer prognostic model with OC consistency. The prognostic value of angiogenesis and immune-related gene expression was evaluated through Kaplan-Meier survival analysis, and the impact of immune infiltration on immune function in ovarian cancer patients was assessed. Functional pathways were identified using the Gene Set Enrichment Analysis (GSEA) method, and the infiltration abundance of immune and stromal components was inferred using the single-sample Gene Set Enrichment Analysis (ssGSEA) method. The influence of angiogenesis on the cellular level of Ovarian Cancer (OC) was explored in single- cell sequencing data, followed by in vitro cell experiments for further validation. The effect of the angiogenesis model on OC was evaluated through the above-mentioned research and experiments, aiming to investigate the mechanism of targeted therapy strategies in ovarian cancer.

Results: Immune-related data were collected from ovarian cancer patients in this study. Through WGCNA analysis, the MEturquoise module was identified, and a total of 1018 hub genes were determined. A prediction model was constructed using machine learning, with CoxBoost+StepCox selected as the best model, leading to the identification of 10 genes associated with ovarian cancer. Patients with high AIDPS had shorter survival time, and GSEA analysis revealed enrichment in immune-related pathways. Single-sample gene set enrichment analysis demonstrated increased immune cell infiltration and malignant stromal changes in the high AIDPS group. Results from in vitro cell experiments showed that silencing RPL31 inhibited the proliferation and migration of ovarian cancer cells while enhancing immune response capability.

Conclusion: AIDPS holds significant clinical significance in Ovarian Cancer (OC) with poor progn

背景:卵巢癌是全球最常见的妇科恶性肿瘤之一:卵巢癌是全球最常见的妇科恶性肿瘤之一,近年来,免疫疗法已成为一种前景广阔的治疗策略。然而,免疫疗法的有效性往往受到免疫逃逸机制的限制:为揭示卵巢癌的免疫应答机制,本研究旨在综合运用加权基因共表达网络分析(WGCNA)、机器学习和单细胞测序分析等方法,系统研究卵巢癌患者免疫浸润相关的分子特征,并通过实验验证免疫应答的分子机制。这项研究可为卵巢癌的免疫疗法提供新的理论基础和治疗策略:方法:从公共数据库中收集相关的卵巢癌数据集。使用 ConsensusCluster- Plus 和 ggplot2 R 软件包对免疫浸润相关基因进行降维和聚类分析。研究人员采用了多种算法来选择具有 OC 一致性的最佳卵巢癌预后模型。通过 Kaplan-Meier 生存分析评估了血管生成和免疫相关基因表达的预后价值,并评估了免疫浸润对卵巢癌患者免疫功能的影响。利用基因组富集分析(Gene Set Enrichment Analysis,GSEA)方法确定了功能通路,并利用单样本基因组富集分析(ssGSEA)方法推断了免疫和基质成分的浸润丰度。在单细胞测序数据中探讨了血管生成对卵巢癌(OC)细胞水平的影响,然后进行体外细胞实验进一步验证。通过上述研究和实验,评估了血管生成模型对卵巢癌的影响,旨在研究卵巢癌靶向治疗策略的机制:本研究收集了卵巢癌患者的免疫相关数据。通过WGCNA分析,确定了MEturquoise模块,并确定了1018个枢纽基因。利用机器学习构建了一个预测模型,其中CoxBoost+StepCox被选为最佳模型,从而确定了10个与卵巢癌相关的基因。高AIDPS患者的生存时间较短,GSEA分析显示了免疫相关通路的富集。单样本基因组富集分析显示,高AIDPS组的免疫细胞浸润和恶性基质变化增加。体外细胞实验结果显示,沉默 RPL31 可抑制卵巢癌细胞的增殖和迁移,同时增强免疫反应能力:结论:AIDPS 在卵巢癌(OC)中具有重要的临床意义,高 AIDPS 患者的预后较差。这些患者表现出更明显的基因组变异、更密集的免疫细胞浸润以及对免疫疗法更强的耐受性。重要的是,抑制 AIDPS 的关键成分 RPL31 的表达可以显著抑制卵巢癌细胞的增殖、迁移和侵袭性,同时刺激效应 T 细胞的细胞毒性并促进免疫反应,从而减缓卵巢癌的进展。
{"title":"Identification of Immune Infiltration-related Molecular Features in Ovarian Cancer Patients and Experimental Validation of Immune Response Molecular Mechanisms through Integrated WGCNA, Machine Learning, and Single-cell Sequencing Analysis.","authors":"Juan Yang, Chengli Wen, Ping Li, Mingxiao Yao, Jing Wang","doi":"10.2174/0115748928297769240611055258","DOIUrl":"https://doi.org/10.2174/0115748928297769240611055258","url":null,"abstract":"<p><strong>Background: </strong>Ovarian cancer is one of the most common gynecological malignancies globally, and immunotherapy has emerged as a promising treatment strategy in recent years. However, the effectiveness of immunotherapy is often limited by immune escape mechanisms.</p><p><strong>Objective: </strong>To unravel the immune response mechanisms in ovarian cancer, this study aimed to employ integrated Weighted Gene Co-expression Network Analysis (WGCNA), machine learning, and single-- cell sequencing analysis to systematically investigate immune infiltration-related molecular features in ovarian cancer patients and experimentally validate the molecular mechanisms of the immune response. This research may provide a new theoretical foundation and treatment strategy for immune-based therapies in ovarian cancer.</p><p><strong>Methods: </strong>Relevant ovarian cancer datasets were collected from public databases. The ConsensusCluster- Plus and ggplot2 R packages were used to perform dimensionality reduction and clustering analysis of immune infiltration-related genes. Various algorithms were employed to select the best ovarian cancer prognostic model with OC consistency. The prognostic value of angiogenesis and immune-related gene expression was evaluated through Kaplan-Meier survival analysis, and the impact of immune infiltration on immune function in ovarian cancer patients was assessed. Functional pathways were identified using the Gene Set Enrichment Analysis (GSEA) method, and the infiltration abundance of immune and stromal components was inferred using the single-sample Gene Set Enrichment Analysis (ssGSEA) method. The influence of angiogenesis on the cellular level of Ovarian Cancer (OC) was explored in single- cell sequencing data, followed by in vitro cell experiments for further validation. The effect of the angiogenesis model on OC was evaluated through the above-mentioned research and experiments, aiming to investigate the mechanism of targeted therapy strategies in ovarian cancer.</p><p><strong>Results: </strong>Immune-related data were collected from ovarian cancer patients in this study. Through WGCNA analysis, the MEturquoise module was identified, and a total of 1018 hub genes were determined. A prediction model was constructed using machine learning, with CoxBoost+StepCox selected as the best model, leading to the identification of 10 genes associated with ovarian cancer. Patients with high AIDPS had shorter survival time, and GSEA analysis revealed enrichment in immune-related pathways. Single-sample gene set enrichment analysis demonstrated increased immune cell infiltration and malignant stromal changes in the high AIDPS group. Results from in vitro cell experiments showed that silencing RPL31 inhibited the proliferation and migration of ovarian cancer cells while enhancing immune response capability.</p><p><strong>Conclusion: </strong>AIDPS holds significant clinical significance in Ovarian Cancer (OC) with poor progn","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141736225","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Withdrawn: Fumarate-1 Mediates the Regulation of Mitochondrial Homeostasis by PGC-1α to Promote Cell Pyroptosis and Inhibit the Thyroid Cancer 富马酸-1通过PGC-1α调控线粒体稳态促进细胞凋亡并抑制甲状腺癌的进展
Pub Date : 2024-07-11 DOI: 10.2174/0115748928282936240530105434
Xiaomei Meng, Dong You, Ruizhen Ren

Since the authors are not responding to the editor’s requests to fulfill the editorial requirement, therefore, the article has been withdrawn.

Bentham Science apologizes to the readers of the journal for any inconvenience this may have caused.

The Bentham Editorial Policy on Article Withdrawal can be found at https://benthamscience.com/editorial-policies-main.php.

Bentham science disclaimer: It is a condition of publication that manuscripts submitted to this journal have not been published and will not be simultaneously submitted or published elsewhere. Furthermore, any data, illustration, structure or table that has been published elsewhere must be reported, and copyright permission for reproduction must be obtained. Plagiarism is strictly forbidden, and by submitting the article for publication the authors agree that the publishers have the legal right to take appropriate action against the authors, if plagiarism or fabricated information is discovered. By submitting a manuscript the authors agree that the copyright of their article is transferred to the publishers if and when the article is accepted for publication.

背景:甲状腺癌是一种罕见的癌症,但在全球范围内发病率越来越高。甲状腺癌的发生和发展与线粒体不稳定性有关,线粒体不稳定性是指线粒体的结构、功能和能量状态发生改变。这些改变导致线粒体代谢失衡,造成细胞损伤和凋亡。然而,线粒体不稳定性与甲状腺癌的分子机制仍鲜为人知:方法:本研究培养了人类甲状腺乳头状癌细胞系(TPC-1 和 K-1)和正常甲状腺细胞系(Nthy-ori 3-1)。分别用oveRNA-FH1和oveRNA-NC转染TPC-1细胞和K-1细胞,分别称为oveRNA-FH1组、oveRNA-NC组、TPC-1组和Nthy-ori 3-1组。实验采用多种方法评估细胞活力、增殖能力、侵袭和迁移能力以及线粒体形态变化和相关因子的表达。这些实验的目的是评估 FH1 对线粒体不稳定性的影响,并阐明甲状腺癌和线粒体不稳定性的具体机制:研究结果表明,与正常甲状腺细胞系相比,FH1在甲状腺乳头状癌细胞系中的表达明显下调。过表达 FH1 会降低 TPC-1 细胞的存活率并抑制细胞增殖率。此外,FH1 的过表达还抑制了细胞的侵袭和迁移能力。在 TPC-1 和 K-1 细胞中观察到异常的线粒体形态变化,而 FH1 过表达则导致线粒体相对正常。过表达 FH1 还会影响融合基因和裂变基因的表达,促进甲状腺癌细胞的裂变,抑制融合。此外,FH1 的过表达还导致炎症和裂解增加。这些结论在体外肿瘤形成实验中得到了进一步验证:结论:FH1通过PGC-1α依赖途径调节线粒体稳态,从而影响热凋亡和细胞凋亡,促进甲状腺癌的进展。
{"title":"Withdrawn: Fumarate-1 Mediates the Regulation of Mitochondrial Homeostasis by PGC-1α to Promote Cell Pyroptosis and Inhibit the Thyroid Cancer","authors":"Xiaomei Meng, Dong You, Ruizhen Ren","doi":"10.2174/0115748928282936240530105434","DOIUrl":"10.2174/0115748928282936240530105434","url":null,"abstract":"<p><p>Since the authors are not responding to the editor’s requests to fulfill the editorial requirement, therefore, the article has been withdrawn.</p><p><p>Bentham Science apologizes to the readers of the journal for any inconvenience this may have caused.</p><p><p>The Bentham Editorial Policy on Article Withdrawal can be found at https://benthamscience.com/editorial-policies-main.php.</p><p><strong>Bentham science disclaimer: </strong>It is a condition of publication that manuscripts submitted to this journal have not been published and will not be simultaneously submitted or published elsewhere. Furthermore, any data, illustration, structure or table that has been published elsewhere must be reported, and copyright permission for reproduction must be obtained. Plagiarism is strictly forbidden, and by submitting the article for publication the authors agree that the publishers have the legal right to take appropriate action against the authors, if plagiarism or fabricated information is discovered. By submitting a manuscript the authors agree that the copyright of their article is transferred to the publishers if and when the article is accepted for publication.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141592507","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tyrosine Kinase Inhibitor Lenvatinib Causes Cardiotoxicity by Inducing Endoplasmic Reticulum Stress and Apoptosis through Activating ATF6, IRE1α and PERK Signaling Pathways. 酪氨酸激酶抑制剂伦伐替尼通过激活 ATF6、IRE1α 和 PERK 信号通路诱导内质网应激和细胞凋亡,从而导致心脏毒性。
Pub Date : 2024-07-11 DOI: 10.2174/0115748928265981231204044653
Siqi Wang, Fang Ji, Xiaoli Gao, Zhiyi Li, Si Lv, Juan Zhang, Jiarui Luo, Dan Li, Jie Yan, Huayang Zhang, Kaicheng Fang, Lin Wu, Miaoling Li

Background: Lenvatinib is a tyrosine kinase inhibitor that can improve progression-free survival in patients with thyroid cancer and hepatocellular carcinoma. However, it is limited by adverse cardiovascular events, including hypertension and cardiac dysfunction. Activation of endoplasmic reticulum stress is involved in cardiomyocyte apoptosis.

Objective: This study aimed to confirm whether the cardiotoxicity of lenvatinib is associated with endoplasmic reticulum stress by targeting the activating transcription factor 6 (ATF6), inositol- requiring enzyme 1α (IRE1α) and protein kinase RNA-like ER kinase (PERK) signaling pathways.

Methods: Male C57/BL6 mice were intragastric administration with 30 mg/kg/day lenvatinib. Electrocardiography (ECG) and echocardiography were used to detect arrhythmias and cardiac function. Neonatal rat cardiomyocytes were treated with lenvatinib for 48h. Cell counting kit (CCK8), 2´,7´-dichlorodihydrofluoresceine diacetate (H2DCFHDA), Hoechst 33258 and dihydrorhodamine 123 were respectively used for evaluating cell viability, the level of reactive oxygen species (ROS), nuclear morphological changes and mitochondrial membrane potential (MMP) level.

Results: Lenvatinib remarkably decreased the posterior wall thickness of left ventricle during diastole and systole but caused little decrease to the left ventricular ejection fraction (LVEF, %). Furthermore, lenvatinib greatly prolonged the corrected QT interval (QTc) and altered the morphology of cardiomyocytes. No dramatic difference in fibrosis was found in mouse cardiac slices. Lenvatinib upregulates apoptosis-related protein expression. In addition, lenvatinib increased ERS-related protein expression (GRP78, CHOP, and ATF6) and enhanced PERK phosphorylation. In neonatal rat cardiac myocytes, lenvatinib markedly decreased the viability of cardiomyocytes and induced apoptosis. Furthermore, ROS production increased and MMP decreased. Similar to the mice experiment, lenvatinib caused upregulation of apoptosis-related and ERS-related proteins and increased the phosphorylation levels of PERK and IRE1α.

Conclusion: Lenvatinib-induced cardiotoxicity is associated with ERS-induced apoptosis by targeting the ATF6, IRE1α, and PERK signaling pathways.

背景介绍伦伐替尼是一种酪氨酸激酶抑制剂,可改善甲状腺癌和肝细胞癌患者的无进展生存期。然而,它也受到不良心血管事件的限制,包括高血压和心功能不全。激活内质网应激参与心肌细胞凋亡:本研究旨在通过靶向激活转录因子6(ATF6)、肌醇需要酶1α(IRE1α)和蛋白激酶RNA样ER激酶(PERK)信号通路,证实来伐替尼的心脏毒性是否与内质网应激有关:雄性C57/BL6小鼠胃内注射30毫克/千克/天的来伐替尼。采用心电图(ECG)和超声心动图检测心律失常和心脏功能。新生大鼠心肌细胞接受来伐替尼治疗48小时。细胞计数试剂盒(CCK8)、2´,7´-二氯二氢荧光素二乙酸酯(H2DCFHDA)、Hoechst 33258和二氢霍达明123分别用于评价细胞活力、活性氧(ROS)水平、核形态变化和线粒体膜电位(MMP)水平:结果:来伐替尼显著降低了左心室舒张期和收缩期的后壁厚度,但对左心室射血分数(LVEF,%)的影响很小。此外,来伐替尼大大延长了校正QT间期(QTc),并改变了心肌细胞的形态。在小鼠心脏切片中未发现纤维化的显著差异。来伐替尼可上调细胞凋亡相关蛋白的表达。此外,来伐替尼还能增加ERS相关蛋白(GRP78、CHOP和ATF6)的表达,并增强PERK的磷酸化。在新生大鼠心肌细胞中,来伐替尼明显降低了心肌细胞的存活率并诱导细胞凋亡。此外,ROS生成增加,MMP减少。与小鼠实验类似,来伐替尼导致凋亡相关蛋白和ERS相关蛋白上调,并增加了PERK和IRE1α的磷酸化水平:结论:来伐替尼诱导的心脏毒性与ERS通过靶向ATF6、IRE1α和PERK信号通路诱导的细胞凋亡有关。
{"title":"Tyrosine Kinase Inhibitor Lenvatinib Causes Cardiotoxicity by Inducing Endoplasmic Reticulum Stress and Apoptosis through Activating ATF6, IRE1α and PERK Signaling Pathways.","authors":"Siqi Wang, Fang Ji, Xiaoli Gao, Zhiyi Li, Si Lv, Juan Zhang, Jiarui Luo, Dan Li, Jie Yan, Huayang Zhang, Kaicheng Fang, Lin Wu, Miaoling Li","doi":"10.2174/0115748928265981231204044653","DOIUrl":"https://doi.org/10.2174/0115748928265981231204044653","url":null,"abstract":"<p><strong>Background: </strong>Lenvatinib is a tyrosine kinase inhibitor that can improve progression-free survival in patients with thyroid cancer and hepatocellular carcinoma. However, it is limited by adverse cardiovascular events, including hypertension and cardiac dysfunction. Activation of endoplasmic reticulum stress is involved in cardiomyocyte apoptosis.</p><p><strong>Objective: </strong>This study aimed to confirm whether the cardiotoxicity of lenvatinib is associated with endoplasmic reticulum stress by targeting the activating transcription factor 6 (ATF6), inositol- requiring enzyme 1α (IRE1α) and protein kinase RNA-like ER kinase (PERK) signaling pathways.</p><p><strong>Methods: </strong>Male C57/BL6 mice were intragastric administration with 30 mg/kg/day lenvatinib. Electrocardiography (ECG) and echocardiography were used to detect arrhythmias and cardiac function. Neonatal rat cardiomyocytes were treated with lenvatinib for 48h. Cell counting kit (CCK8), 2´,7´-dichlorodihydrofluoresceine diacetate (H2DCFHDA), Hoechst 33258 and dihydrorhodamine 123 were respectively used for evaluating cell viability, the level of reactive oxygen species (ROS), nuclear morphological changes and mitochondrial membrane potential (MMP) level.</p><p><strong>Results: </strong>Lenvatinib remarkably decreased the posterior wall thickness of left ventricle during diastole and systole but caused little decrease to the left ventricular ejection fraction (LVEF, %). Furthermore, lenvatinib greatly prolonged the corrected QT interval (QTc) and altered the morphology of cardiomyocytes. No dramatic difference in fibrosis was found in mouse cardiac slices. Lenvatinib upregulates apoptosis-related protein expression. In addition, lenvatinib increased ERS-related protein expression (GRP78, CHOP, and ATF6) and enhanced PERK phosphorylation. In neonatal rat cardiac myocytes, lenvatinib markedly decreased the viability of cardiomyocytes and induced apoptosis. Furthermore, ROS production increased and MMP decreased. Similar to the mice experiment, lenvatinib caused upregulation of apoptosis-related and ERS-related proteins and increased the phosphorylation levels of PERK and IRE1α.</p><p><strong>Conclusion: </strong>Lenvatinib-induced cardiotoxicity is associated with ERS-induced apoptosis by targeting the ATF6, IRE1α, and PERK signaling pathways.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141592508","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epigallocatechin Gallate Derivative Y6 Reverses Oxaliplatin Resistance in Hepatocellular Carcinoma via Targeting the MiR-338-3p/HIF-1α/TWIST Axis to Inhibit EMT. 表没食子儿茶素没食子酸酯衍生物 Y6 通过靶向 MiR-338-3p/HIF-1α/TWIST 轴抑制 EMT 逆转肝细胞癌的奥沙利铂耐药性
Pub Date : 2024-07-11 DOI: 10.2174/0115748928293750240619092747
Chuan Huang, Si-Hong Wang, Tao-Tao Liu, Mei-Yi Wu, Kai-Ning Cai, Zhan-Hong Xie, Rui-Qiang Zhao, Yan Wen

Background: The emergence of drug resistance to oxaliplatin (OXA) is one of the critical obstacles in the therapy of advanced Hepatocellular Carcinoma (HCC). As an ethyl derivative of the natural compound epigallocatechin gallate (epigallocatechin-3-gallate, EGCG), Y6 was found to be able to enhance the sensitivity of HCC cells to doxorubicin. This study aimed to investigate the effect of Y6 on oxaliplatin resistance in HCC.

Methods: MTT was used to determine the reversal effect of Y6 on OXA resistance. To further explore the reversal mechanism, we treated OXA alone or in combination with Y6 or EGCG in drugresistant cells and observed the morphological changes of the cells. At the same time, transwell assay was used to detect the invasion and migration ability of cells. Moreover, Real-time PCR and Western blot analysis were performed to determine the expression levels of the miR-338-3p gene, HIF-1α/Twist proteins, and EMT-related proteins.

Results: We found that Y6 could inhibit the proliferation of HCC cells and effectively reverse the drug resistance of oxaliplatin-resistant human liver cancer cells (SMMC-7721/OXA) to OXA, and the reversal effect was more significant than that of its lead drug EGCG. Most of the cells in the control group and OXA group showed typical mesenchymal-like cell morphology, while most of the cells in co-administration groups showed typical epithelioid cell morphology, and the ability of the cells to invade and migrate decreased dramatically, particularly in Y6 plus OXA group. At the same time, Y6 could up-regulate the EMT epithelial marker protein E-cadherin and down-regulate the interstitial marker protein Vimentin. In addition, in co-administration groups, the expression of miR-338-3p was up-regulated, while the expression of HIF-1α and Twist was down-regulated.

Conclusion: Y6 significantly enhanced the susceptibility of drug-resistant cells to OXA, and the process may be related to the regulation of miR-338-3p/HIF-1α / TWIST pathway to inhibit EMT. Therefore, Y6 could be considered an effective medication resistance reversal agent, which could improve the therapeutic effect for hepatocellular cancer patients.

背景:奥沙利铂(OXA)耐药性的出现是晚期肝细胞癌(HCC)治疗的关键障碍之一。作为天然化合物表没食子儿茶素没食子酸酯(表没食子儿茶素-3-没食子酸酯,EGCG)的乙基衍生物,Y6被发现能够提高HCC细胞对多柔比星的敏感性。本研究旨在探讨 Y6 对 HCC 奥沙利铂耐药性的影响:方法:采用 MTT 测定 Y6 对奥沙利铂耐药性的逆转作用。为了进一步探讨逆转机制,我们对耐药细胞单独或与 Y6 或 EGCG 联合处理 OXA,并观察细胞的形态学变化。同时,我们还采用了Transwell试验来检测细胞的侵袭和迁移能力。此外,还进行了实时 PCR 和 Western 印迹分析,以确定 miR-338-3p 基因、HIF-1α/Twist 蛋白和 EMT 相关蛋白的表达水平:结果:我们发现Y6能抑制HCC细胞的增殖,并有效逆转奥沙利铂耐药人肝癌细胞(SMMC-7721/OXA)对OXA的耐药性,其逆转效果比其主药EGCG更显著。对照组和OXA组的大部分细胞呈现典型的间质样细胞形态,而联合给药组的大部分细胞呈现典型的上皮样细胞形态,细胞的侵袭和迁移能力显著下降,尤其是Y6加OXA组。同时,Y6 能上调 EMT 上皮标志蛋白 E-cadherin,下调间质标志蛋白 Vimentin。此外,在联合用药组中,miR-338-3p的表达上调,而HIF-1α和Twist的表达下调:结论:Y6能明显增强耐药细胞对OXA的敏感性,这一过程可能与调控miR-338-3p/HIF-1α/TWIST通路抑制EMT有关。因此,Y6 可被视为一种有效的耐药性逆转剂,可提高肝癌患者的治疗效果。
{"title":"Epigallocatechin Gallate Derivative Y6 Reverses Oxaliplatin Resistance in Hepatocellular Carcinoma via Targeting the MiR-338-3p/HIF-1α/TWIST Axis to Inhibit EMT.","authors":"Chuan Huang, Si-Hong Wang, Tao-Tao Liu, Mei-Yi Wu, Kai-Ning Cai, Zhan-Hong Xie, Rui-Qiang Zhao, Yan Wen","doi":"10.2174/0115748928293750240619092747","DOIUrl":"https://doi.org/10.2174/0115748928293750240619092747","url":null,"abstract":"<p><strong>Background: </strong>The emergence of drug resistance to oxaliplatin (OXA) is one of the critical obstacles in the therapy of advanced Hepatocellular Carcinoma (HCC). As an ethyl derivative of the natural compound epigallocatechin gallate (epigallocatechin-3-gallate, EGCG), Y6 was found to be able to enhance the sensitivity of HCC cells to doxorubicin. This study aimed to investigate the effect of Y6 on oxaliplatin resistance in HCC.</p><p><strong>Methods: </strong>MTT was used to determine the reversal effect of Y6 on OXA resistance. To further explore the reversal mechanism, we treated OXA alone or in combination with Y6 or EGCG in drugresistant cells and observed the morphological changes of the cells. At the same time, transwell assay was used to detect the invasion and migration ability of cells. Moreover, Real-time PCR and Western blot analysis were performed to determine the expression levels of the miR-338-3p gene, HIF-1α/Twist proteins, and EMT-related proteins.</p><p><strong>Results: </strong>We found that Y6 could inhibit the proliferation of HCC cells and effectively reverse the drug resistance of oxaliplatin-resistant human liver cancer cells (SMMC-7721/OXA) to OXA, and the reversal effect was more significant than that of its lead drug EGCG. Most of the cells in the control group and OXA group showed typical mesenchymal-like cell morphology, while most of the cells in co-administration groups showed typical epithelioid cell morphology, and the ability of the cells to invade and migrate decreased dramatically, particularly in Y6 plus OXA group. At the same time, Y6 could up-regulate the EMT epithelial marker protein E-cadherin and down-regulate the interstitial marker protein Vimentin. In addition, in co-administration groups, the expression of miR-338-3p was up-regulated, while the expression of HIF-1α and Twist was down-regulated.</p><p><strong>Conclusion: </strong>Y6 significantly enhanced the susceptibility of drug-resistant cells to OXA, and the process may be related to the regulation of miR-338-3p/HIF-1α / TWIST pathway to inhibit EMT. Therefore, Y6 could be considered an effective medication resistance reversal agent, which could improve the therapeutic effect for hepatocellular cancer patients.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-07-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141592506","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Radiotherapy Enhancing and Radioprotective Properties of Berberine: A Systematic Review. 小檗碱的放疗增强和放射保护特性:系统综述。
Pub Date : 2024-07-09 DOI: 10.2174/0115748928315442240624120104
Elham Raeisi, Saeid Heidari-Soureshjani, Catherine Mt Sherwin, Zeinab Bagheri

Background: Natural compounds such as Berberine (Ber) have been considered due to favorable anticancer properties, low side effects, and availability along with chemotherapy treatments.

Objectives: This study aimed to investigate the radiosensitizing and radioprotective properties of Ber.

Methods: In this systematic review that was performed according to PRISMA 2020 guidelines, we searched the publications before 25 Sep 2023 in Web of Science, PubMed, Scopus, Embase, and Cochrane Library databases. After determining inclusion and exclusion criteria, data were extracted and imported into an Excel form, and the results of the studies were reviewed.

Results: Ber by reducing the levels of reactive oxygen species (ROS), malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-α), transforming growth factor-beta 1 (TGF-β1), and increasing interleukin 10 (IL-10) levels, showed its antioxidant and anti-inflammatory properties against ionizing radiation. Reducing cell cytotoxicity and apoptosis were other radioprotective properties of Ber. Conversely, in cancer cells, Ber, via inducing oxidative stress and accumulation ROS in tumor tissues, inducing DNA damage, mitochondrial dysfunction and hyperpolarization, inducing apoptosis, and cell cycle arrest, inhibits the up-regulation of hypoxia-inducible factor-1 alpha (HIF-1α) and vascular endothelial growth factor (VEGF) revealed radiosensitizing properties.

Conclusion: Ber, via various mechanisms, showed favorable radioprotective and radiosensitizing properties in clinical and experimental studies. However, more clinical studies are needed in this field.

背景:小檗碱(Berberine,Ber)等天然化合物因其良好的抗癌特性、低副作用以及可与化疗药物同时使用而被认为是抗癌药物:本研究旨在探讨小檗碱的放射增敏和放射保护特性:本系统性综述根据 PRISMA 2020 指南进行,我们在 Web of Science、PubMed、Scopus、Embase 和 Cochrane Library 数据库中检索了 2023 年 9 月 25 日之前的出版物。确定纳入和排除标准后,提取数据并导入Excel表格,对研究结果进行回顾:Ber 通过降低活性氧(ROS)、丙二醛(MDA)、肿瘤坏死因子-α(TNF-α)、转化生长因子-β1(TGF-β1)的水平,以及提高白细胞介素 10(IL-10)的水平,显示了其抗氧化和抗炎特性。减少细胞毒性和细胞凋亡是 Ber 的其他辐射保护特性。相反,在癌细胞中,Ber 通过诱导氧化应激和肿瘤组织中 ROS 的积累,诱导 DNA 损伤、线粒体功能障碍和超极化,诱导细胞凋亡和细胞周期停滞,抑制缺氧诱导因子-1α(HIF-1α)和血管内皮生长因子(VEGF)的上调,显示出其放射致敏特性:结论:在临床和实验研究中,Ber 通过各种机制显示出良好的放射保护和放射致敏特性。然而,这一领域还需要更多的临床研究。
{"title":"Radiotherapy Enhancing and Radioprotective Properties of Berberine: A Systematic Review.","authors":"Elham Raeisi, Saeid Heidari-Soureshjani, Catherine Mt Sherwin, Zeinab Bagheri","doi":"10.2174/0115748928315442240624120104","DOIUrl":"https://doi.org/10.2174/0115748928315442240624120104","url":null,"abstract":"<p><strong>Background: </strong>Natural compounds such as Berberine (Ber) have been considered due to favorable anticancer properties, low side effects, and availability along with chemotherapy treatments.</p><p><strong>Objectives: </strong>This study aimed to investigate the radiosensitizing and radioprotective properties of Ber.</p><p><strong>Methods: </strong>In this systematic review that was performed according to PRISMA 2020 guidelines, we searched the publications before 25 Sep 2023 in Web of Science, PubMed, Scopus, Embase, and Cochrane Library databases. After determining inclusion and exclusion criteria, data were extracted and imported into an Excel form, and the results of the studies were reviewed.</p><p><strong>Results: </strong>Ber by reducing the levels of reactive oxygen species (ROS), malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-α), transforming growth factor-beta 1 (TGF-β1), and increasing interleukin 10 (IL-10) levels, showed its antioxidant and anti-inflammatory properties against ionizing radiation. Reducing cell cytotoxicity and apoptosis were other radioprotective properties of Ber. Conversely, in cancer cells, Ber, via inducing oxidative stress and accumulation ROS in tumor tissues, inducing DNA damage, mitochondrial dysfunction and hyperpolarization, inducing apoptosis, and cell cycle arrest, inhibits the up-regulation of hypoxia-inducible factor-1 alpha (HIF-1α) and vascular endothelial growth factor (VEGF) revealed radiosensitizing properties.</p><p><strong>Conclusion: </strong>Ber, via various mechanisms, showed favorable radioprotective and radiosensitizing properties in clinical and experimental studies. However, more clinical studies are needed in this field.</p>","PeriodicalId":94186,"journal":{"name":"Recent patents on anti-cancer drug discovery","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-07-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141565466","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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
Recent patents on anti-cancer drug discovery
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