首页 > 最新文献

Medical Oncology最新文献

英文 中文
A phase-I study of second-line S-IROX for unresectable pancreatic cancer after gemcitabine plus nab-paclitaxel failure. 吉西他滨+纳布-紫杉醇治疗失败后二线S-IROX治疗不可切除胰腺癌的I期研究。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-07-05 DOI: 10.1007/s12032-024-02438-x
Mitsuru Okuno, Tsuyoshi Mukai, Keisuke Iwata, Akihiro Takagi, Yuki Ito, Yosuke Ohashi, Ryuichi Tezuka, Yuhei Iwasa, Shota Iwata, Eiichi Tomita

Gemcitabine plus nab-paclitaxel (GnP) and FOLFIRINOX are widely used as first-line regimens for unresectable pancreatic cancer (PC). When GnP therapy is selected, considering patient age or condition, second-line FOLFIRINOX is sometimes difficult to administer owing to its toxicity. This study aimed to determine the recommended dose (RD) of S-IROX (S-1, oxaliplatin, and irinotecan combination) regimens in patients with unresectable PC after first-line GnP failure. This phase-I study used the "3 + 3" dose-escalation design with two dose levels. Patients who failed first-line GnP therapy for unresectable PC were enrolled. Oxaliplatin and irinotecan were administered on day 1, and S-1 was administered orally twice daily on days 1-7, followed by 7 days of rest. The primary endpoints were dose-limiting toxicities (DLTs) and determination of RD. The secondary endpoint was the evaluation of potential antitumor activity. Nine patients received the second-line S-IROX regimen. In level-0 (S-1, 80 mg/m2; oxaliplatin, 85 mg/m2; and irinotecan, 120 mg/m2), no patient experienced DLT; however, one patient experienced grade 3 neutropenia. At level-1 (irinotecan increased to 150 mg/m2), one of six patients experienced DLTs, including G3 diarrhea. The RD was confirmed at the level-1 dose. The response rate, disease control rate, median progression-free survival, and median overall survival were 33.3%, 77.8%, 172 (range:77-422) days, and 414 (101-685) days, respectively. One patient underwent surgery after the second-line S-IROX therapy. Second-line S-IROX treatment was deemed acceptable. The RD was set at level-1 dose (S-1, 80 mg/m2; oxaliplatin, 85 mg/m2; and irinotecan, 150 mg/m2).

吉西他滨+纳布-紫杉醇(GnP)和FOLFIRINOX被广泛用作不可切除胰腺癌(PC)的一线治疗方案。在选择GnP疗法时,考虑到患者的年龄或病情,二线FOLFIRINOX有时因其毒性而难以施用。本研究旨在确定S-IROX(S-1、奥沙利铂和伊立替康联合疗法)方案在一线GnP治疗失败后不可切除的PC患者中的推荐剂量(RD)。这项 I 期研究采用 "3 + 3 "剂量递增设计,有两个剂量水平。研究对象为GnP一线治疗失败的不可切除PC患者。第1天服用奥沙利铂和伊立替康,第1-7天口服S-1,每天两次,然后休息7天。主要终点是剂量限制性毒性(DLT)和RD测定。次要终点是评估潜在的抗肿瘤活性。九名患者接受了二线 S-IROX 方案。在 0 级方案(S-1,80 毫克/平方米;奥沙利铂,85 毫克/平方米;伊立替康,120 毫克/平方米)中,没有患者出现 DLT;但有一名患者出现 3 级中性粒细胞减少。在 1 级疗法(伊立替康增至 150 毫克/平方米)中,六名患者中有一人出现了 DLT,包括 G3 腹泻。RD在1级剂量时得到确认。反应率、疾病控制率、中位无进展生存期和中位总生存期分别为33.3%、77.8%、172(范围:77-422)天和414(101-685)天。一名患者在二线 S-IROX 治疗后接受了手术。二线 S-IROX 治疗被认为是可以接受的。RD 设定为一级剂量(S-1,80 毫克/平方米;奥沙利铂,85 毫克/平方米;伊立替康,150 毫克/平方米)。
{"title":"A phase-I study of second-line S-IROX for unresectable pancreatic cancer after gemcitabine plus nab-paclitaxel failure.","authors":"Mitsuru Okuno, Tsuyoshi Mukai, Keisuke Iwata, Akihiro Takagi, Yuki Ito, Yosuke Ohashi, Ryuichi Tezuka, Yuhei Iwasa, Shota Iwata, Eiichi Tomita","doi":"10.1007/s12032-024-02438-x","DOIUrl":"10.1007/s12032-024-02438-x","url":null,"abstract":"<p><p>Gemcitabine plus nab-paclitaxel (GnP) and FOLFIRINOX are widely used as first-line regimens for unresectable pancreatic cancer (PC). When GnP therapy is selected, considering patient age or condition, second-line FOLFIRINOX is sometimes difficult to administer owing to its toxicity. This study aimed to determine the recommended dose (RD) of S-IROX (S-1, oxaliplatin, and irinotecan combination) regimens in patients with unresectable PC after first-line GnP failure. This phase-I study used the \"3 + 3\" dose-escalation design with two dose levels. Patients who failed first-line GnP therapy for unresectable PC were enrolled. Oxaliplatin and irinotecan were administered on day 1, and S-1 was administered orally twice daily on days 1-7, followed by 7 days of rest. The primary endpoints were dose-limiting toxicities (DLTs) and determination of RD. The secondary endpoint was the evaluation of potential antitumor activity. Nine patients received the second-line S-IROX regimen. In level-0 (S-1, 80 mg/m<sup>2</sup>; oxaliplatin, 85 mg/m<sup>2</sup>; and irinotecan, 120 mg/m<sup>2</sup>), no patient experienced DLT; however, one patient experienced grade 3 neutropenia. At level-1 (irinotecan increased to 150 mg/m<sup>2</sup>), one of six patients experienced DLTs, including G3 diarrhea. The RD was confirmed at the level-1 dose. The response rate, disease control rate, median progression-free survival, and median overall survival were 33.3%, 77.8%, 172 (range:77-422) days, and 414 (101-685) days, respectively. One patient underwent surgery after the second-line S-IROX therapy. Second-line S-IROX treatment was deemed acceptable. The RD was set at level-1 dose (S-1, 80 mg/m<sup>2</sup>; oxaliplatin, 85 mg/m<sup>2</sup>; and irinotecan, 150 mg/m<sup>2</sup>).</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-07-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141534731","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular pathway of anticancer effect of next-generation HSP90 inhibitors XL-888 and Debio0932 in neuroblastoma cell line. 下一代 HSP90 抑制剂 XL-888 和 Debio0932 在神经母细胞瘤细胞系中的抗癌作用的分子途径。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-07-03 DOI: 10.1007/s12032-024-02428-z
Özlem Kaplan, Nazan Gökşen Tosun

Neuroblastoma is a common nervous system tumor in childhood, and current treatments are not adequate. HSP90 is a molecular chaperone protein that plays a critical role in the regulation of cancer-related proteins. HSP90 inhibition may exert anticancer effects by targeting cancer-related processes such as tumor growth, cell proliferation, metastasis, and apoptosis. Therefore, HSP90 inhibition is a promising strategy in the treatment of various types of cancer, and the development of next-generation inhibitors could potentially lead to more effective and safer treatments. XL-888 and Debio0932 is a next-generation HSP90 inhibitor and can inhibit the correct folding and stabilization of client proteins that cancer-associated HSP90 helps to fold correctly. In this study, we aimed to investigate the comprehensive molecular pathways of the anticancer activity of XL-888 and Debio0932 in human neuroblastoma cells SH-SY5Y. The cytotoxic effects of XL-888 and Debio0932 on the neuroblastoma cell line SH-SY5Y cells were evaluated by MTT assay. Then, the effect of these HSP90 inhibitors on the expression of important genes in cancer was revealed by Quantitative Real Time Polymerase Chain Reaction (qRT-PCR) method. The qRT-PCR data were evaluated using Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) biological process tools. Finally, the effect of HSP90 inhibitors on HSP27, HSP70 and HSP90 protein expression was investigated by Western blotting analysis. The results revealed that XL-888 and Debio0932 had a role in regulating many cancer-related pathways such as migration, invasion, metastasis, angiogenesis, and apoptosis in SH-SY5Y cells. In conclusion, it shows that HSP90 inhibitors can be considered as a promising candidate in the treatment of neuroblastoma and resistance to chemotherapy.

神经母细胞瘤是儿童时期常见的神经系统肿瘤,目前的治疗方法并不完善。HSP90 是一种分子伴侣蛋白,在调控癌症相关蛋白方面发挥着关键作用。抑制 HSP90 可针对肿瘤生长、细胞增殖、转移和凋亡等癌症相关过程发挥抗癌作用。因此,HSP90 抑制剂是治疗各种类型癌症的一种很有前景的策略,下一代抑制剂的开发有可能带来更有效、更安全的治疗方法。XL-888 和 Debio0932 是下一代 HSP90 抑制剂,可抑制癌症相关 HSP90 帮助正确折叠和稳定的客户蛋白。本研究旨在探讨 XL-888 和 Debio0932 在人神经母细胞瘤细胞 SH-SY5Y 中抗癌活性的综合分子途径。研究采用MTT法评估了XL-888和Debio0932对神经母细胞瘤细胞株SH-SY5Y细胞的细胞毒性作用。然后,通过定量实时聚合酶链式反应(qRT-PCR)方法揭示了这些 HSP90 抑制剂对癌症重要基因表达的影响。利用京都基因组百科全书(KEGG)和基因本体(GO)生物过程工具对qRT-PCR数据进行了评估。最后,通过 Western 印迹分析研究了 HSP90 抑制剂对 HSP27、HSP70 和 HSP90 蛋白表达的影响。结果显示,XL-888和Debio0932在调控SH-SY5Y细胞的迁移、侵袭、转移、血管生成和凋亡等多种癌症相关通路中发挥作用。综上所述,HSP90抑制剂在治疗神经母细胞瘤和化疗耐药性方面具有广阔的前景。
{"title":"Molecular pathway of anticancer effect of next-generation HSP90 inhibitors XL-888 and Debio0932 in neuroblastoma cell line.","authors":"Özlem Kaplan, Nazan Gökşen Tosun","doi":"10.1007/s12032-024-02428-z","DOIUrl":"10.1007/s12032-024-02428-z","url":null,"abstract":"<p><p>Neuroblastoma is a common nervous system tumor in childhood, and current treatments are not adequate. HSP90 is a molecular chaperone protein that plays a critical role in the regulation of cancer-related proteins. HSP90 inhibition may exert anticancer effects by targeting cancer-related processes such as tumor growth, cell proliferation, metastasis, and apoptosis. Therefore, HSP90 inhibition is a promising strategy in the treatment of various types of cancer, and the development of next-generation inhibitors could potentially lead to more effective and safer treatments. XL-888 and Debio0932 is a next-generation HSP90 inhibitor and can inhibit the correct folding and stabilization of client proteins that cancer-associated HSP90 helps to fold correctly. In this study, we aimed to investigate the comprehensive molecular pathways of the anticancer activity of XL-888 and Debio0932 in human neuroblastoma cells SH-SY5Y. The cytotoxic effects of XL-888 and Debio0932 on the neuroblastoma cell line SH-SY5Y cells were evaluated by MTT assay. Then, the effect of these HSP90 inhibitors on the expression of important genes in cancer was revealed by Quantitative Real Time Polymerase Chain Reaction (qRT-PCR) method. The qRT-PCR data were evaluated using Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) biological process tools. Finally, the effect of HSP90 inhibitors on HSP27, HSP70 and HSP90 protein expression was investigated by Western blotting analysis. The results revealed that XL-888 and Debio0932 had a role in regulating many cancer-related pathways such as migration, invasion, metastasis, angiogenesis, and apoptosis in SH-SY5Y cells. In conclusion, it shows that HSP90 inhibitors can be considered as a promising candidate in the treatment of neuroblastoma and resistance to chemotherapy.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11222184/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141492579","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","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 ZNF334 in cervical cancer: implications for EMT reversal and tumor suppression. ZNF334 在宫颈癌中的作用:对 EMT 逆转和肿瘤抑制的影响
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-07-02 DOI: 10.1007/s12032-024-02433-2
Qian Li, Xiangyi Zhou, Jiayi Xiao, Yijia Gong, Xue Gong, Bianfei Shao, Jianhua Wang, Lijuan Zhao, Qi Xiong, Yue Wu, Jun Tang, Qiyu Yang, Junying Tang, Tingxiu Xiang

Zinc-finger proteins are involved in many biological processes. However, the role of Zinc-finger protein 334 (ZNF334) in cervical cancer remains unidentified. This study showed that promoter methylation of ZNF334 was responsible for its reduced expression. ZNF334 suppressed malignant biological behaviors in cervical cancer. Notably, ZNF334 reversed the EMT process both in vitro and in vivo. RNA-seq coupled with bioinformatics analysis caught P3H3 which is upregulated by ZNF334. Dual-luciferase reporter and Chromatin immunoprecipitation assays illustrated that ZNF334 directly regulate P3H3. Knockdown of P3H3 attenuated the reversal of EMT induced by ZNF334. Additionally, ZNF334 overexpression sensitized cervical cancer cells to the cytotoxic effects of paclitaxel, cyclosporine and sunitinib. In conclusions, this study illustrated that DNA methylation-based silencing ZNF334 played a vital role in cervical cancer, by regulating P3H3 in turn affects EMT. ZNF334 has the potential to become a novel diagnostic biomarker and a potential treatment target for cervical cancer.

锌指蛋白参与了许多生物过程。然而,锌指蛋白 334(ZNF334)在宫颈癌中的作用仍未确定。这项研究表明,ZNF334启动子甲基化是其表达减少的原因。ZNF334抑制了宫颈癌的恶性生物学行为。值得注意的是,ZNF334在体外和体内都能逆转EMT过程。RNA-seq结合生物信息学分析发现了ZNF334上调的P3H3。双荧光素酶报告和染色质免疫沉淀分析表明,ZNF334 直接调控 P3H3。敲除 P3H3 可减轻 ZNF334 诱导的 EMT 的逆转。此外,ZNF334 的过表达使宫颈癌细胞对紫杉醇、环孢素和舒尼替尼的细胞毒性作用敏感。总之,这项研究表明,基于 DNA 甲基化的 ZNF334 沉默在宫颈癌中发挥了重要作用,它通过调节 P3H3 进而影响 EMT。ZNF334有望成为宫颈癌的新型诊断生物标志物和潜在治疗靶点。
{"title":"Role of ZNF334 in cervical cancer: implications for EMT reversal and tumor suppression.","authors":"Qian Li, Xiangyi Zhou, Jiayi Xiao, Yijia Gong, Xue Gong, Bianfei Shao, Jianhua Wang, Lijuan Zhao, Qi Xiong, Yue Wu, Jun Tang, Qiyu Yang, Junying Tang, Tingxiu Xiang","doi":"10.1007/s12032-024-02433-2","DOIUrl":"10.1007/s12032-024-02433-2","url":null,"abstract":"<p><p>Zinc-finger proteins are involved in many biological processes. However, the role of Zinc-finger protein 334 (ZNF334) in cervical cancer remains unidentified. This study showed that promoter methylation of ZNF334 was responsible for its reduced expression. ZNF334 suppressed malignant biological behaviors in cervical cancer. Notably, ZNF334 reversed the EMT process both in vitro and in vivo. RNA-seq coupled with bioinformatics analysis caught P3H3 which is upregulated by ZNF334. Dual-luciferase reporter and Chromatin immunoprecipitation assays illustrated that ZNF334 directly regulate P3H3. Knockdown of P3H3 attenuated the reversal of EMT induced by ZNF334. Additionally, ZNF334 overexpression sensitized cervical cancer cells to the cytotoxic effects of paclitaxel, cyclosporine and sunitinib. In conclusions, this study illustrated that DNA methylation-based silencing ZNF334 played a vital role in cervical cancer, by regulating P3H3 in turn affects EMT. ZNF334 has the potential to become a novel diagnostic biomarker and a potential treatment target for cervical cancer.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141492580","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Comment on "Focused cancer pathway analysis revealed unique therapeutic targets in retinoblastoma". 就 "聚焦癌症通路分析揭示视网膜母细胞瘤的独特治疗靶点 "发表评论
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-07-02 DOI: 10.1007/s12032-024-02445-y
Hinpetch Daungsupawong, Viroj Wiwanitkit
{"title":"Comment on \"Focused cancer pathway analysis revealed unique therapeutic targets in retinoblastoma\".","authors":"Hinpetch Daungsupawong, Viroj Wiwanitkit","doi":"10.1007/s12032-024-02445-y","DOIUrl":"10.1007/s12032-024-02445-y","url":null,"abstract":"","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141492542","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CTLA-4 silencing could promote anti-tumor effects in hepatocellular. 沉默CTLA-4可促进肝细胞抗肿瘤作用
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-07-02 DOI: 10.1007/s12032-024-02361-1
Amirhossein Mardi, Mahsan Alizadeh, Amir Shahabaddin Abdolalizadeh, Amir Baghbanzadeh, Behzad Baradaran, Ali Aghebaqti-Maleki, Siamak Sandoghchian Shotorbani, Mohammad Movloudi, Leili Aghebati-Maleki

Preclinical and clinical research showed that immune checkpoint blockade provides beneficial effects for many patients with liver cancer. This study aimed to assess the effect of CTLA-4-specific siRNA on the proliferation, cell cycle, migration, and apoptosis of HePG2 cells. Transfection of siRNA was performed by electroporation. The viability of cells was determined through MTT assay. Flow cytometry was performed to investigate the cell cycle and apoptosis rate, and the wound-healing assay was used to determine HepG2 cells migration. The expression levels of CTLA-4, c-Myc, Ki-67, BCL-2, BAX, caspase-9 (CAS9), and MMP-2,9,13 were measured by qRT-PCR. Transfection of specific CTLA-4-siRNA significantly inhibited the expression of the CTLA-4 gene. Also, our results revealed that CTLA-4 silencing diminished the proliferation and migration as well as induced the apoptosis of HePG2 cells. CTLA-4-siRNA transfection induced the cell cycle arrest in G2 phase. Moreover, CTLA-4-siRNA transfection reduced the expression levels of c-Myc, Ki-67, BCL-2, MMP-2,9,13, and elevated the expression levels of BAX and caspase-9. Our results suggest that silencing CTLA-4 through specific siRNA may be a promising strategy for future therapeutic interventions for treating liver cancer.

临床前和临床研究表明,免疫检查点阻断可为许多肝癌患者带来益处。本研究旨在评估 CTLA-4 特异性 siRNA 对 HePG2 细胞增殖、细胞周期、迁移和凋亡的影响。siRNA 的转染采用电穿孔法。细胞活力通过 MTT 试验测定。流式细胞仪检测细胞周期和凋亡率,伤口愈合试验检测 HepG2 细胞的迁移。qRT-PCR检测了CTLA-4、c-Myc、Ki-67、BCL-2、BAX、caspase-9(CAS9)和MMP-2、9、13的表达水平。转染特异性 CTLA-4-siRNA 能明显抑制 CTLA-4 基因的表达。同时,我们的研究结果表明,沉默 CTLA-4 可减少 HePG2 细胞的增殖和迁移,并诱导其凋亡。CTLA-4-siRNA 转染诱导细胞周期停滞在 G2 期。此外,CTLA-4-siRNA转染降低了c-Myc、Ki-67、BCL-2、MMP-2、9、13的表达水平,并升高了BAX和caspase-9的表达水平。我们的研究结果表明,通过特异性 siRNA 沉默 CTLA-4 可能是未来治疗肝癌的一种有前途的干预策略。
{"title":"CTLA-4 silencing could promote anti-tumor effects in hepatocellular.","authors":"Amirhossein Mardi, Mahsan Alizadeh, Amir Shahabaddin Abdolalizadeh, Amir Baghbanzadeh, Behzad Baradaran, Ali Aghebaqti-Maleki, Siamak Sandoghchian Shotorbani, Mohammad Movloudi, Leili Aghebati-Maleki","doi":"10.1007/s12032-024-02361-1","DOIUrl":"10.1007/s12032-024-02361-1","url":null,"abstract":"<p><p>Preclinical and clinical research showed that immune checkpoint blockade provides beneficial effects for many patients with liver cancer. This study aimed to assess the effect of CTLA-4-specific siRNA on the proliferation, cell cycle, migration, and apoptosis of HePG2 cells. Transfection of siRNA was performed by electroporation. The viability of cells was determined through MTT assay. Flow cytometry was performed to investigate the cell cycle and apoptosis rate, and the wound-healing assay was used to determine HepG2 cells migration. The expression levels of CTLA-4, c-Myc, Ki-67, BCL-2, BAX, caspase-9 (CAS9), and MMP-2,9,13 were measured by qRT-PCR. Transfection of specific CTLA-4-siRNA significantly inhibited the expression of the CTLA-4 gene. Also, our results revealed that CTLA-4 silencing diminished the proliferation and migration as well as induced the apoptosis of HePG2 cells. CTLA-4-siRNA transfection induced the cell cycle arrest in G2 phase. Moreover, CTLA-4-siRNA transfection reduced the expression levels of c-Myc, Ki-67, BCL-2, MMP-2,9,13, and elevated the expression levels of BAX and caspase-9. Our results suggest that silencing CTLA-4 through specific siRNA may be a promising strategy for future therapeutic interventions for treating liver cancer.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141492543","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The role of histamine and its receptors in breast cancer: from pathology to therapeutic targets. 组胺及其受体在乳腺癌中的作用:从病理学到治疗目标。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-07-01 DOI: 10.1007/s12032-024-02437-y
Hossein Azimi, Afifeh Jafari, Mahafarin Maralani, Homa Davoodi

Breast cancer is the most common malignancy in women, and despite the development of new treatment methods and the decreasing mortality rate in recent years, one of the clinical problems in breast cancer treatment is chronic inflammation in the tumor microenvironment. Histamine, an inflammatory mediator, is produced by tumor cells and can induce chronic inflammation and the growth of some tumors by recruiting inflammatory cells. It can also affect tumor physiopathology, antitumor treatment efficiency, and patient survival. Antihistamines, as histamine receptor antagonists, play a role in modulating the effects of these receptors in tumor cells and can affect some treatment methods for breast cancer therapy; in this review, we investigate the role of histamine, its receptors, and antihistamines in breast cancer pathology and treatment methods.

乳腺癌是女性最常见的恶性肿瘤,尽管近年来新的治疗方法层出不穷,死亡率也在不断下降,但肿瘤微环境中的慢性炎症仍是乳腺癌治疗的临床难题之一。组胺是一种炎症介质,由肿瘤细胞产生,可通过招募炎症细胞诱发慢性炎症和某些肿瘤的生长。它还会影响肿瘤的生理病理、抗肿瘤治疗效率和患者生存。抗组胺药作为组胺受体拮抗剂,在调节肿瘤细胞中这些受体的作用方面发挥着作用,并可影响乳腺癌治疗的一些治疗方法;在这篇综述中,我们探讨了组胺、组胺受体和抗组胺药在乳腺癌病理和治疗方法中的作用。
{"title":"The role of histamine and its receptors in breast cancer: from pathology to therapeutic targets.","authors":"Hossein Azimi, Afifeh Jafari, Mahafarin Maralani, Homa Davoodi","doi":"10.1007/s12032-024-02437-y","DOIUrl":"10.1007/s12032-024-02437-y","url":null,"abstract":"<p><p>Breast cancer is the most common malignancy in women, and despite the development of new treatment methods and the decreasing mortality rate in recent years, one of the clinical problems in breast cancer treatment is chronic inflammation in the tumor microenvironment. Histamine, an inflammatory mediator, is produced by tumor cells and can induce chronic inflammation and the growth of some tumors by recruiting inflammatory cells. It can also affect tumor physiopathology, antitumor treatment efficiency, and patient survival. Antihistamines, as histamine receptor antagonists, play a role in modulating the effects of these receptors in tumor cells and can affect some treatment methods for breast cancer therapy; in this review, we investigate the role of histamine, its receptors, and antihistamines in breast cancer pathology and treatment methods.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141476935","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Which drug would be more effective in treating patients with leptomeningeal metastases from breast cancer: intrathecal thiotepa or methotrexate? 鞘内注射硫替派和甲氨蝶呤,哪种药物治疗乳腺癌脑膜转移患者更有效?
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-06-29 DOI: 10.1007/s12032-024-02439-w
Kadri Altundag
{"title":"Which drug would be more effective in treating patients with leptomeningeal metastases from breast cancer: intrathecal thiotepa or methotrexate?","authors":"Kadri Altundag","doi":"10.1007/s12032-024-02439-w","DOIUrl":"10.1007/s12032-024-02439-w","url":null,"abstract":"","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-06-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141476936","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The role of IL-17 in the pathogenesis and treatment of glioblastoma-an update on the state of the art and future perspectives. IL-17在胶质母细胞瘤发病机制和治疗中的作用--最新进展和未来展望。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-06-25 DOI: 10.1007/s12032-024-02434-1
Dariusz Łaszczych, Aleksandra Czernicka, Karol Gostomczyk, Łukasz Szylberg, Jędrzej Borowczak

Glioblastoma (GBM) is the most common malignant brain tumor, which, despite significant progress made in the last years in the field of neuro-oncology, remains an incurable disease. GBM has a poor prognosis with a median survival of 12-15 months, and its aggressive clinical course is related to rapid growth, extensive infiltration of adjacent tissues, resistance to chemotherapy, radiotherapy and immunotherapy, and frequent relapse. Currently, several molecular biomarkers are used in clinical practice to predict patient prognosis and response to treatment. However, due to the overall unsatisfactory efficacy of standard multimodal treatment and the remaining poor prognosis, there is an urgent need for new biomarkers and therapeutic strategies for GBM. Recent evidence suggests that GBM tumorigenesis is associated with crosstalk between cancer, immune and stromal cells mediated by various cytokines. One of the key factors involved in this process appears to be interleukin-17 (IL-17), a pro-inflammatory cytokine that is significantly upregulated in the serum and tissue of GBM patients. IL-17 plays a key role in tumorigenesis, angiogenesis, and recurrence of GBM by activating pro-oncogenic signaling pathways and promoting cell survival, proliferation, and invasion. IL-17 facilitates the immunomodulation of the tumor microenvironment by promoting immune cells infiltration and cytokine secretion. In this article we review the latest scientific reports to provide an update on the role of IL-17 role in tumorigenesis, tumor microenvironment, diagnosis, prognosis, and treatment of GBM.

胶质母细胞瘤(GBM)是最常见的恶性脑肿瘤,尽管近年来在神经肿瘤学领域取得了重大进展,但它仍然是一种不治之症。GBM 的预后较差,中位生存期仅为 12-15 个月,其侵袭性临床病程与生长迅速、广泛浸润邻近组织、对化疗、放疗和免疫治疗耐药以及频繁复发有关。目前,临床上使用了多种分子生物标志物来预测患者的预后和对治疗的反应。然而,由于标准多模式治疗的总体疗效并不理想,预后仍然很差,因此迫切需要针对 GBM 的新生物标志物和治疗策略。最近的证据表明,GBM 肿瘤的发生与各种细胞因子介导的癌症、免疫细胞和基质细胞之间的串扰有关。参与这一过程的关键因素之一似乎是白细胞介素-17(IL-17),这是一种促炎细胞因子,在 GBM 患者的血清和组织中显著上调。IL-17 通过激活促癌信号通路并促进细胞存活、增殖和侵袭,在 GBM 的肿瘤发生、血管生成和复发过程中发挥着关键作用。IL-17 通过促进免疫细胞浸润和细胞因子分泌,推动肿瘤微环境的免疫调节。本文回顾了最新的科学报道,介绍了 IL-17 在 GBM 的肿瘤发生、肿瘤微环境、诊断、预后和治疗中的作用。
{"title":"The role of IL-17 in the pathogenesis and treatment of glioblastoma-an update on the state of the art and future perspectives.","authors":"Dariusz Łaszczych, Aleksandra Czernicka, Karol Gostomczyk, Łukasz Szylberg, Jędrzej Borowczak","doi":"10.1007/s12032-024-02434-1","DOIUrl":"10.1007/s12032-024-02434-1","url":null,"abstract":"<p><p>Glioblastoma (GBM) is the most common malignant brain tumor, which, despite significant progress made in the last years in the field of neuro-oncology, remains an incurable disease. GBM has a poor prognosis with a median survival of 12-15 months, and its aggressive clinical course is related to rapid growth, extensive infiltration of adjacent tissues, resistance to chemotherapy, radiotherapy and immunotherapy, and frequent relapse. Currently, several molecular biomarkers are used in clinical practice to predict patient prognosis and response to treatment. However, due to the overall unsatisfactory efficacy of standard multimodal treatment and the remaining poor prognosis, there is an urgent need for new biomarkers and therapeutic strategies for GBM. Recent evidence suggests that GBM tumorigenesis is associated with crosstalk between cancer, immune and stromal cells mediated by various cytokines. One of the key factors involved in this process appears to be interleukin-17 (IL-17), a pro-inflammatory cytokine that is significantly upregulated in the serum and tissue of GBM patients. IL-17 plays a key role in tumorigenesis, angiogenesis, and recurrence of GBM by activating pro-oncogenic signaling pathways and promoting cell survival, proliferation, and invasion. IL-17 facilitates the immunomodulation of the tumor microenvironment by promoting immune cells infiltration and cytokine secretion. In this article we review the latest scientific reports to provide an update on the role of IL-17 role in tumorigenesis, tumor microenvironment, diagnosis, prognosis, and treatment of GBM.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11199243/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141450861","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Genipin's potential as an anti-cancer agent: from phytochemical origins to clinical prospects. 吉尼平作为抗癌剂的潜力:从植物化学起源到临床前景。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-06-25 DOI: 10.1007/s12032-024-02429-y
Lapava Natallia, Aida Dama, Era Gorica, Karaliova Darya, Sheila I Peña-Corona, Hernán Cortés, Antonello Santini, Dietrich Büsselberg, Gerardo Leyva-Gómez, Javad Sharifi-Rad

This comprehensive review delves into the multifaceted aspects of genipin, a bioactive compound derived from medicinal plants, focusing on its anti-cancer potential. The review begins by detailing the sources and phytochemical properties of genipin, underscoring its significance in traditional medicine and its transition into contemporary cancer research. It then explores the intricate relationship between genipin's chemical structure and its observed anti-cancer activity, highlighting the molecular underpinnings contributing to its therapeutic potential. This is complemented by a thorough analysis of preclinical studies, which investigates genipin's efficacy against various cancer cell lines and its mechanisms of action at the cellular level. A crucial component of the review is the examination of genipin's bioavailability and pharmacokinetics, providing insights into how the compound is absorbed, distributed, metabolized, and excreted in the body. Then, this review offers a general and updated overview of the anti-cancer studies of genipin and its derivatives based on its basic molecular mechanisms, induction of apoptosis, inhibition of cell proliferation, and disruption of cancer cell signaling pathways. We include information that complements the genipin study, such as toxicity data, and we differentiate this review by including commercial status, disposition, and regulation. Also, this review of genipin stands out for incorporating information on proposals for a technological approach through its load in nanotechnology to improve its bioavailability. The culmination of this information positions genipin as a promising candidate for developing novel anti-cancer drugs capable of supplementing or enhancing current cancer therapies.

吉尼平是一种从药用植物中提取的生物活性化合物,本综述从多方面深入探讨了吉尼平的抗癌潜力。综述首先详细介绍了吉尼平的来源和植物化学特性,强调了它在传统医学中的重要性,以及它在当代癌症研究中的转变。然后探讨了基尼肽的化学结构与所观察到的抗癌活性之间错综复杂的关系,强调了其治疗潜力的分子基础。此外,还对临床前研究进行了深入分析,研究了吉尼平对各种癌症细胞系的疗效及其在细胞水平上的作用机制。本综述的一个重要组成部分是对基尼平的生物利用度和药代动力学的研究,深入探讨了该化合物在体内的吸收、分布、代谢和排泄过程。然后,本综述根据基尼平及其衍生物的基本分子机制、诱导细胞凋亡、抑制细胞增殖和破坏癌细胞信号传导途径,对基尼平及其衍生物的抗癌研究进行了全面和最新的概述。我们纳入了对吉尼平研究起补充作用的信息,如毒性数据,并通过纳入商业状况、处置和监管信息使本综述与众不同。此外,这篇关于吉尼平的综述还特别纳入了有关通过在纳米技术中添加吉尼平来提高其生物利用率的技术方法建议的信息。综合这些信息,吉尼平有望成为开发新型抗癌药物的候选药物,以补充或增强目前的癌症疗法。
{"title":"Genipin's potential as an anti-cancer agent: from phytochemical origins to clinical prospects.","authors":"Lapava Natallia, Aida Dama, Era Gorica, Karaliova Darya, Sheila I Peña-Corona, Hernán Cortés, Antonello Santini, Dietrich Büsselberg, Gerardo Leyva-Gómez, Javad Sharifi-Rad","doi":"10.1007/s12032-024-02429-y","DOIUrl":"10.1007/s12032-024-02429-y","url":null,"abstract":"<p><p>This comprehensive review delves into the multifaceted aspects of genipin, a bioactive compound derived from medicinal plants, focusing on its anti-cancer potential. The review begins by detailing the sources and phytochemical properties of genipin, underscoring its significance in traditional medicine and its transition into contemporary cancer research. It then explores the intricate relationship between genipin's chemical structure and its observed anti-cancer activity, highlighting the molecular underpinnings contributing to its therapeutic potential. This is complemented by a thorough analysis of preclinical studies, which investigates genipin's efficacy against various cancer cell lines and its mechanisms of action at the cellular level. A crucial component of the review is the examination of genipin's bioavailability and pharmacokinetics, providing insights into how the compound is absorbed, distributed, metabolized, and excreted in the body. Then, this review offers a general and updated overview of the anti-cancer studies of genipin and its derivatives based on its basic molecular mechanisms, induction of apoptosis, inhibition of cell proliferation, and disruption of cancer cell signaling pathways. We include information that complements the genipin study, such as toxicity data, and we differentiate this review by including commercial status, disposition, and regulation. Also, this review of genipin stands out for incorporating information on proposals for a technological approach through its load in nanotechnology to improve its bioavailability. The culmination of this information positions genipin as a promising candidate for developing novel anti-cancer drugs capable of supplementing or enhancing current cancer therapies.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141450860","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A drug repurposing study identifies novel FOXM1 inhibitors with in vitro activity against breast cancer cells. 一项药物再利用研究发现了对乳腺癌细胞具有体外活性的新型 FOXM1 抑制剂。
IF 2.8 4区 医学 Q2 ONCOLOGY Pub Date : 2024-06-25 DOI: 10.1007/s12032-024-02427-0
Khaled A N Abusharkh, Ferah Comert Onder, Venhar Çınar, Zuhal Hamurcu, Bulent Ozpolat, Mehmet Ay

FOXM1, a proto-oncogenic transcription factor, plays a critical role in cancer development and treatment resistance in cancers, particularly in breast cancer. Thus, this study aimed to identify potential FOXM1 inhibitors through computational screening of drug databases, followed by in vitro validation of their inhibitory activity against breast cancer cells. In silico studies involved pharmacophore modeling using the FOXM1 inhibitor, FDI-6, followed by virtual screening of DrugBank and Selleckchem databases. The selected drugs were prepared for molecular docking, and the crystal structure of FOXM1 was pre-processed for docking simulations. In vitro studies included MTT assays to assess cytotoxicity, and Western blot analysis to evaluate protein expression levels. Our study identified Pantoprazole and Rabeprazole as potential FOXM1 inhibitors through in silico screening and molecular docking. Molecular dynamics simulations confirmed stable interactions of these drugs with FOXM1. In vitro experiments showed both Pantoprazole and Rabeprazole exhibited strong FOXM1 inhibition at effective concentrations and that showed inhibition of cell proliferation. Rabeprazole showed the inhibitor activity at 10 µM in BT-20 and MCF-7 cell lines. Pantoprazole exhibited FOXM1 inhibition at 30 µM and in BT-20 cells and at 70 µM in MCF-7 cells, respectively. Our current study provides the first evidence that Rabeprazole and Pantoprazole can bind to FOXM1 and inhibit its activity and downstream signaling, including eEF2K and pEF2, in breast cancer cells. These findings indicate that rabeprazole and pantoprazole inhibit FOXM1 and breast cancer cell proliferation, and they can be used for FOXM1-targeted therapy in breast or other cancers driven by FOXM1.

FOXM1是一种原癌基因转录因子,在癌症尤其是乳腺癌的发展和耐药性中起着关键作用。因此,本研究旨在通过对药物数据库进行计算筛选,找出潜在的 FOXM1 抑制剂,然后对其对乳腺癌细胞的抑制活性进行体外验证。硅学研究包括使用 FOXM1 抑制剂 FDI-6 进行药效学建模,然后对 DrugBank 和 Selleckchem 数据库进行虚拟筛选。选定的药物已准备好进行分子对接,FOXM1 的晶体结构也已进行了对接模拟的预处理。体外研究包括评估细胞毒性的 MTT 试验和评估蛋白质表达水平的 Western 印迹分析。我们的研究通过硅学筛选和分子对接确定了泮托拉唑和雷贝拉唑为潜在的 FOXM1 抑制剂。分子动力学模拟证实了这些药物与 FOXM1 的稳定相互作用。体外实验显示,泮托拉唑和雷贝拉唑在有效浓度下均表现出很强的 FOXM1 抑制作用,并能抑制细胞增殖。在 BT-20 和 MCF-7 细胞系中,雷贝拉唑在 10 µM 的浓度下显示出抑制活性。泮托拉唑对 BT-20 细胞的 FOXM1 抑制浓度为 30 µM,对 MCF-7 细胞的抑制浓度为 70 µM。我们目前的研究首次证明,雷贝拉唑和泮托拉唑能与 FOXM1 结合,并抑制其活性和下游信号转导,包括乳腺癌细胞中的 eEF2K 和 pEF2。这些研究结果表明,雷贝拉唑和泮托拉唑能抑制 FOXM1 和乳腺癌细胞的增殖,可用于乳腺癌或其他由 FOXM1 驱动的癌症的 FOXM1 靶向治疗。
{"title":"A drug repurposing study identifies novel FOXM1 inhibitors with in vitro activity against breast cancer cells.","authors":"Khaled A N Abusharkh, Ferah Comert Onder, Venhar Çınar, Zuhal Hamurcu, Bulent Ozpolat, Mehmet Ay","doi":"10.1007/s12032-024-02427-0","DOIUrl":"10.1007/s12032-024-02427-0","url":null,"abstract":"<p><p>FOXM1, a proto-oncogenic transcription factor, plays a critical role in cancer development and treatment resistance in cancers, particularly in breast cancer. Thus, this study aimed to identify potential FOXM1 inhibitors through computational screening of drug databases, followed by in vitro validation of their inhibitory activity against breast cancer cells. In silico studies involved pharmacophore modeling using the FOXM1 inhibitor, FDI-6, followed by virtual screening of DrugBank and Selleckchem databases. The selected drugs were prepared for molecular docking, and the crystal structure of FOXM1 was pre-processed for docking simulations. In vitro studies included MTT assays to assess cytotoxicity, and Western blot analysis to evaluate protein expression levels. Our study identified Pantoprazole and Rabeprazole as potential FOXM1 inhibitors through in silico screening and molecular docking. Molecular dynamics simulations confirmed stable interactions of these drugs with FOXM1. In vitro experiments showed both Pantoprazole and Rabeprazole exhibited strong FOXM1 inhibition at effective concentrations and that showed inhibition of cell proliferation. Rabeprazole showed the inhibitor activity at 10 µM in BT-20 and MCF-7 cell lines. Pantoprazole exhibited FOXM1 inhibition at 30 µM and in BT-20 cells and at 70 µM in MCF-7 cells, respectively. Our current study provides the first evidence that Rabeprazole and Pantoprazole can bind to FOXM1 and inhibit its activity and downstream signaling, including eEF2K and pEF2, in breast cancer cells. These findings indicate that rabeprazole and pantoprazole inhibit FOXM1 and breast cancer cell proliferation, and they can be used for FOXM1-targeted therapy in breast or other cancers driven by FOXM1.</p>","PeriodicalId":18433,"journal":{"name":"Medical Oncology","volume":null,"pages":null},"PeriodicalIF":2.8,"publicationDate":"2024-06-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11199234/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141450859","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","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
Medical Oncology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号: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