首页 > 最新文献

Journal of Medicinal Chemistry最新文献

英文 中文
3-Deazaneplanocin A (DZNep): A Drug That Deserves a Second Look. 3-Deazaneplanocin A (DZNep):值得重新审视的药物。
IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-11 DOI: 10.1021/acs.jmedchem.4c01566
Victor E Marquez

The emerging data compiled during the past five years on 3-deazaneplanocin (DZNep) provide compelling evidence to reevaluate this drug as a better alternative over the specific catalytic inhibitors of histone methyl transferases (HTMs). The indirect mechanism of DZNep via inhibition of AdoHcy-ase, once considered a liability due to possible side effects, has now shown to be rather beneficial as additional pathways targeted by DZNep are important contributors to its superior anticancer properties. Furthermore, DZNep has demonstrated the ability to induce proteasomal degradation of its target and reduce toxicity in combination with well-established antitumor therapies in animal models. In addition, DZNep has shown important effects in suppressing fibrosis and inflammation in liver, kidney, peritoneum, and airways. Finally, inhibition of mRNA m6A methylation by DZNep suppresses the synthesis of the viral genome in SARS-Cov-2 infection and promises to have important therapeutic value when combined with its potent antiviral efficacy and anti-inflammatory effects.

过去五年中有关 3-去氮杂环庚烷霉素(DZNep)的新数据为重新评估这种药物提供了令人信服的证据,使其成为组蛋白甲基转移酶(HTMs)特异性催化抑制剂的更好替代品。DZNep 通过抑制 AdoHcy-ase 的间接机制曾因可能产生的副作用而被认为是一种负担,但现在却被证明是有益的,因为 DZNep 靶向的其他途径是其卓越抗癌特性的重要因素。此外,在动物模型中,DZNep 还能诱导蛋白酶体降解其靶点,并与成熟的抗肿瘤疗法联合使用以降低毒性。此外,DZNep 对抑制肝脏、肾脏、腹膜和呼吸道的纤维化和炎症也有重要作用。最后,DZNep 对 mRNA m6A 甲基化的抑制可抑制 SARS-Cov-2 感染中病毒基因组的合成,结合其强大的抗病毒功效和抗炎作用,有望产生重要的治疗价值。
{"title":"3-Deazaneplanocin A (DZNep): A Drug That Deserves a Second Look.","authors":"Victor E Marquez","doi":"10.1021/acs.jmedchem.4c01566","DOIUrl":"10.1021/acs.jmedchem.4c01566","url":null,"abstract":"<p><p>The emerging data compiled during the past five years on 3-deazaneplanocin (DZNep) provide compelling evidence to reevaluate this drug as a better alternative over the specific catalytic inhibitors of histone methyl transferases (HTMs). The indirect mechanism of DZNep via inhibition of AdoHcy-ase, once considered a liability due to possible side effects, has now shown to be rather beneficial as additional pathways targeted by DZNep are important contributors to its superior anticancer properties. Furthermore, DZNep has demonstrated the ability to induce proteasomal degradation of its target and reduce toxicity in combination with well-established antitumor therapies in animal models. In addition, DZNep has shown important effects in suppressing fibrosis and inflammation in liver, kidney, peritoneum, and airways. Finally, inhibition of mRNA m<sup>6</sup>A methylation by DZNep suppresses the synthesis of the viral genome in SARS-Cov-2 infection and promises to have important therapeutic value when combined with its potent antiviral efficacy and anti-inflammatory effects.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-10-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142398713","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery of Novel 5-(Pyridazin-3-yl)pyrimidine-2,4(1H,3H)-dione Derivatives as Potent and Orally Bioavailable Inhibitors Targeting Ecto-5'-nucleotidase. 发现新型 5-(哒嗪-3-基)嘧啶-2,4(1H,3H)-二酮衍生物作为靶向外-5'-核苷酸酶的强效口服生物抑制剂
IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-10 DOI: 10.1021/acs.jmedchem.4c01793
Yu Xu, Dan Liu, Wenzhuang Zhang, Zhining Liu, Jingjing Liu, Wanling Zhang, Rongxing Song, Jia Li, Fan Yang, Yue Wang, Dunkai Liu, Gaofei Qian, Hua Tang, Xijing Chen, Yisheng Lai

Ecto-5-nucleotidase (CD73) is overexpressed in a variety of cancers and associated with the immunosuppressive tumor microenvironment, making it an attractive target for cancer immunotherapy. Herein, we designed and synthesized a series of novel (pyridazine-3-yl)pyrimidine-2,4(1H,3H)-dione derivatives as CD73 inhibitors. These compounds exhibited remarkable inhibitory activity against CD73 in both enzymatic biochemical and cellular assays. Among them, compound 35j proved to be one of the most potent inhibitors and an uncompetitive inhibitor with no obvious cytotoxicity. This compound showed high metabolic stability in rat liver microsomes and favorable pharmacokinetic profiles in rats (T1/2 = 3.37 h, F = 50.24%). Importantly, orally administered 35j significantly inhibited tumor growth in the triple-negative breast cancer 4T1 mouse model (TGI = 73.6%, 50 mg/kg). Immunoassays suggested that 35j remarkably increased the infiltration of positive immune cells, thereby reinvigorating antitumor immunity. These results demonstrate that 35j is a potent CD73 inhibitor worthy of further development.

外向-5-核苷酸酶(CD73)在多种癌症中过度表达,并与免疫抑制性肿瘤微环境相关,因此成为癌症免疫疗法的一个有吸引力的靶点。在此,我们设计并合成了一系列新型(哒嗪-3-基)嘧啶-2,4(1H,3H)-二酮衍生物作为 CD73 抑制剂。这些化合物在酶生化和细胞实验中都表现出了对 CD73 的显著抑制活性。其中,化合物 35j 被证明是最有效的抑制剂之一,也是一种无明显细胞毒性的非竞争性抑制剂。该化合物在大鼠肝脏微粒体中的代谢稳定性很高,在大鼠体内的药代动力学特征良好(T1/2 = 3.37 h,F = 50.24%)。重要的是,口服 35j 能显著抑制三阴性乳腺癌 4T1 小鼠模型的肿瘤生长(TGI = 73.6%,50 mg/kg)。免疫测定表明,35j 能明显增加阳性免疫细胞的浸润,从而重振抗肿瘤免疫力。这些结果表明,35j 是一种有效的 CD73 抑制剂,值得进一步开发。
{"title":"Discovery of Novel 5-(Pyridazin-3-yl)pyrimidine-2,4(1<i>H</i>,3<i>H</i>)-dione Derivatives as Potent and Orally Bioavailable Inhibitors Targeting Ecto-5'-nucleotidase.","authors":"Yu Xu, Dan Liu, Wenzhuang Zhang, Zhining Liu, Jingjing Liu, Wanling Zhang, Rongxing Song, Jia Li, Fan Yang, Yue Wang, Dunkai Liu, Gaofei Qian, Hua Tang, Xijing Chen, Yisheng Lai","doi":"10.1021/acs.jmedchem.4c01793","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01793","url":null,"abstract":"<p><p>Ecto-5-nucleotidase (CD73) is overexpressed in a variety of cancers and associated with the immunosuppressive tumor microenvironment, making it an attractive target for cancer immunotherapy. Herein, we designed and synthesized a series of novel (pyridazine-3-yl)pyrimidine-2,4(1<i>H</i>,3<i>H</i>)-dione derivatives as CD73 inhibitors. These compounds exhibited remarkable inhibitory activity against CD73 in both enzymatic biochemical and cellular assays. Among them, compound <b>35j</b> proved to be one of the most potent inhibitors and an uncompetitive inhibitor with no obvious cytotoxicity. This compound showed high metabolic stability in rat liver microsomes and favorable pharmacokinetic profiles in rats (<i>T</i><sub>1/2</sub> = 3.37 h, <i>F</i> = 50.24%). Importantly, orally administered <b>35j</b> significantly inhibited tumor growth in the triple-negative breast cancer 4T1 mouse model (TGI = 73.6%, 50 mg/kg). Immunoassays suggested that <b>35j</b> remarkably increased the infiltration of positive immune cells, thereby reinvigorating antitumor immunity. These results demonstrate that <b>35j</b> is a potent CD73 inhibitor worthy of further development.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142386400","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery of a Potent, Orally Active, and Long-Lasting P2X7 Receptor Antagonist as a Preclinical Candidate for Delaying the Progression of Chronic Kidney Disease. 发现一种强效、口服活性和长效 P2X7 受体拮抗剂,作为延缓慢性肾病进展的临床前候选药物。
IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-10 Epub Date: 2024-09-23 DOI: 10.1021/acs.jmedchem.4c01395
Ruijia Zhang, Kaiyue Su, Letian Yang, Huaichuan Duan, Lei Tang, Minghai Tang, Min Zhao, Neng Ye, Xiaoying Cai, Xueqin Jiang, Na Li, Jing Peng, Xinlu Zhang, Lingkai Tang, Qiang Qiu, Lijuan Chen, Wenshuang Wu, Jianping Hu, Liang Ma, Haoyu Ye

Chronic kidney disease (CKD) is a condition characterized by functional deterioration with sustained inflammation and progressive fibrosis of the kidneys affecting over 800 million people worldwide. The P2X7 receptor (P2X7R) plays a key role in CKD progression. Our previous P2X7R antagonists demonstrated good efficacy for treating kidney injury but were limited by low oral exposure and short half-life, restricting their application. This study reports the optimization of P2X7R antagonists for better oral pharmacokinetics. The candidate compound 13a with the respective IC50 of 34.86 and 25.28 nM against human and murine P2X7R, administered orally at 10 mg/kg in mice, exhibits a remarkably long half-life of 161.64 h, with a high exposure of 1,163,980.55 μg·h/L. Oral administration of 13a (0.3 or 1.0 mg/kg, twice weekly) significantly reduced renal injury and fibrosis in unilateral ureteral obstruction and adenine diet-induced mice models, highlighting its potential for delaying the progression of CKD.

慢性肾脏病(CKD)是一种以肾脏持续炎症和进行性纤维化导致功能衰退为特征的疾病,影响着全球 8 亿多人。P2X7 受体(P2X7R)在慢性肾脏病的进展中起着关键作用。我们以前的 P2X7R 拮抗剂对治疗肾损伤有很好的疗效,但受限于低口服暴露量和短半衰期,限制了其应用。本研究对 P2X7R 拮抗剂进行了优化,以获得更好的口服药代动力学。候选化合物 13a 对人类和小鼠 P2X7R 的 IC50 分别为 34.86 和 25.28 nM,小鼠口服剂量为 10 mg/kg,半衰期长达 161.64 h,暴露量高达 1,163,980.55 μg-h/L。口服 13a(0.3 或 1.0 毫克/千克,每周两次)可明显减轻单侧输尿管梗阻和腺嘌呤饮食诱导的小鼠模型的肾损伤和肾纤维化,突出了其延缓慢性肾脏病进展的潜力。
{"title":"Discovery of a Potent, Orally Active, and Long-Lasting P2X7 Receptor Antagonist as a Preclinical Candidate for Delaying the Progression of Chronic Kidney Disease.","authors":"Ruijia Zhang, Kaiyue Su, Letian Yang, Huaichuan Duan, Lei Tang, Minghai Tang, Min Zhao, Neng Ye, Xiaoying Cai, Xueqin Jiang, Na Li, Jing Peng, Xinlu Zhang, Lingkai Tang, Qiang Qiu, Lijuan Chen, Wenshuang Wu, Jianping Hu, Liang Ma, Haoyu Ye","doi":"10.1021/acs.jmedchem.4c01395","DOIUrl":"10.1021/acs.jmedchem.4c01395","url":null,"abstract":"<p><p>Chronic kidney disease (CKD) is a condition characterized by functional deterioration with sustained inflammation and progressive fibrosis of the kidneys affecting over 800 million people worldwide. The P2X7 receptor (P2X7R) plays a key role in CKD progression. Our previous P2X7R antagonists demonstrated good efficacy for treating kidney injury but were limited by low oral exposure and short half-life, restricting their application. This study reports the optimization of P2X7R antagonists for better oral pharmacokinetics. The candidate compound <b>13a</b> with the respective IC<sub>50</sub> of 34.86 and 25.28 nM against human and murine P2X7R, administered orally at 10 mg/kg in mice, exhibits a remarkably long half-life of 161.64 h, with a high exposure of 1,163,980.55 μg·h/L. Oral administration of <b>13a</b> (0.3 or 1.0 mg/kg, twice weekly) significantly reduced renal injury and fibrosis in unilateral ureteral obstruction and adenine diet-induced mice models, highlighting its potential for delaying the progression of CKD.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142277338","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Boosting Med Chem Education: Integrating Biology for Drug Discovery Talents. 促进医学化学教育:整合生物学,培养药物发现人才。
IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-10 Epub Date: 2024-09-24 DOI: 10.1021/acs.jmedchem.4c02208
Qiu Sun, Liang Ouyang
{"title":"Boosting Med Chem Education: Integrating Biology for Drug Discovery Talents.","authors":"Qiu Sun, Liang Ouyang","doi":"10.1021/acs.jmedchem.4c02208","DOIUrl":"10.1021/acs.jmedchem.4c02208","url":null,"abstract":"","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142306580","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Computationally Driven Discovery of a BCR-ABL1 Kinase Inhibitor with Activity in Multidrug-Resistant Chronic Myeloid Leukemia. 计算驱动发现对多药耐药慢性髓性白血病有疗效的 BCR-ABL1 激酶抑制剂
IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-10 Epub Date: 2024-09-23 DOI: 10.1021/acs.jmedchem.4c01852
Jarvis Hill, R Houston Givhan, Bin Yi, Robert M Jones, Eugene F Douglass, Yaguang Xi, Henry F Schaefer, David Crich

The permeability glycoprotein, encoded by the ABCB1 gene, is widely implicated in multidrug resistance (MDR), as it has been shown to reduce the intracellular concentration of most small molecule therapeutics, including the majority of the breakpoint cluster region Abelson proto-oncogene 1 (BCR-ABL1) kinase inhibitors used in the treatment of Philadelphia chromosome positive (Ph+) leukemias. With this in mind, we describe an integrated theoretical and experimental approach to shed light on substituent effects in the pendant anilino moiety of 4-anilinoquinazolines and 4-anilinoquinoline-3-carbonitrile-based kinase inhibitors and their influence on P-gp-mediated efflux. This analysis culminated in the identification of a hydroxylamine-bearing, dual cSRC/BCR-ABL1 kinase inhibitor 16a that exhibits a marked reduction in P-gp-mediated efflux ratio and potent activity in a Ph+ patient-derived cell line (K562) and an MDR-Ph+ patient-derived cell line (K562/Dox) overexpressing P-gp. Overall, we demonstrate that the P-gp-mediated efflux ratio can be minimized by computationally driven optimization of the molecular dipole and/or cpKa without recourse to intramolecular hydrogen bonds.

由 ABCB1 基因编码的渗透性糖蛋白被广泛认为与多药耐药性(MDR)有关,因为它已被证明能降低大多数小分子治疗药物的细胞内浓度,包括用于治疗费城染色体阳性(Ph+)白血病的大多数断点集群区阿贝尔森原癌基因 1(BCR-ABL1)激酶抑制剂。有鉴于此,我们介绍了一种理论与实验相结合的方法,以揭示 4-苯胺基喹唑类和 4-苯胺基喹啉-3-甲腈类激酶抑制剂的垂苯胺基取代基效应及其对 P-gp 介导的外流的影响。这一分析最终确定了一种含羟胺的 cSRC/BCR-ABL1 双激酶抑制剂 16a,它能显著降低 P-gp 介导的外流比率,并能在 Ph+ 患者衍生细胞系(K562)和过表达 P-gp 的 MDR-Ph+ 患者衍生细胞系(K562/Dox)中发挥强效活性。总之,我们证明了通过计算驱动优化分子偶极和/或 cpKa 可以最大限度地降低 P-gp 介导的外流率,而无需借助分子内氢键。
{"title":"Computationally Driven Discovery of a BCR-ABL1 Kinase Inhibitor with Activity in Multidrug-Resistant Chronic Myeloid Leukemia.","authors":"Jarvis Hill, R Houston Givhan, Bin Yi, Robert M Jones, Eugene F Douglass, Yaguang Xi, Henry F Schaefer, David Crich","doi":"10.1021/acs.jmedchem.4c01852","DOIUrl":"10.1021/acs.jmedchem.4c01852","url":null,"abstract":"<p><p>The permeability glycoprotein, encoded by the <i>ABCB1</i> gene, is widely implicated in multidrug resistance (MDR), as it has been shown to reduce the intracellular concentration of most small molecule therapeutics, including the majority of the breakpoint cluster region Abelson proto-oncogene 1 (BCR-ABL1) kinase inhibitors used in the treatment of Philadelphia chromosome positive (Ph+) leukemias. With this in mind, we describe an integrated theoretical and experimental approach to shed light on substituent effects in the pendant anilino moiety of 4-anilinoquinazolines and 4-anilinoquinoline-3-carbonitrile-based kinase inhibitors and their influence on P-gp-mediated efflux. This analysis culminated in the identification of a hydroxylamine-bearing, dual cSRC/BCR-ABL1 kinase inhibitor <b>16a</b> that exhibits a marked reduction in P-gp-mediated efflux ratio and potent activity in a Ph+ patient-derived cell line (K562) and an MDR-Ph+ patient-derived cell line (K562/Dox) overexpressing P-gp. Overall, we demonstrate that the P-gp-mediated efflux ratio can be minimized by computationally driven optimization of the molecular dipole and/or cp<i>K</i><sub>a</sub> without recourse to intramolecular hydrogen bonds.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11472320/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142277337","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Discovery of Benzo[d]oxazoles as Novel Dual Small-Molecule Inhibitors Targeting PD-1/PD-L1 and VISTA Pathway 发现以 PD-1/PD-L1 和 VISTA 通路为靶点的苯并[d]恶唑类新型双重小分子抑制剂
IF 7.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-10 DOI: 10.1021/acs.jmedchem.4c01899
Kaizhen Wang, Shi Cai, Yao Cheng, Zhihao Qi, Xiang Ni, Kuojun Zhang, Yibei Xiao, Xiangyu Zhang, Tianyu Wang
The blockers of programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) pathway have achieved great clinical success. However, the limited efficacy and low tumor response rate of anti-PD-1/PD-L1 monotherapy limit the clinical application of PD-1/PD-L1 inhibitors. V-domain immunoglobulin suppressor of T-cell activation (VISTA), a novel checkpoint regulator, exhibits potential synergy with PD-1/PD-L1 in enhancing antitumor immunity. Herein, we report the discovery of benzo[d]oxazole B3 as novel dual small-molecule inhibitors targeting PD-1/PD-L1 and VISTA with high PD-1/PD-L1 inhibitory activity and VISTA binding affinity. B3 rescues the immunosuppression of T-cells mediated by PD-L1 and VISTA and activates antitumor immunity effectively. Moreover, B3 could induce degradation of PD-L1 and VISTA in tumor cell. Furthermore, B3 displays significant in vivo antitumor efficacy in a CT26 mouse model. Our results discover B3 as a promising dual PD-1/PD-L1 and VISTA inhibitor, providing a novel therapeutic strategy to overcome the limitations of current anti-PD-1/PD-L1 therapy.
程序性细胞死亡-1(PD-1)/程序性细胞死亡-配体1(PD-L1)通路阻断剂在临床上取得了巨大成功。然而,抗PD-1/PD-L1单药治疗的有限疗效和较低的肿瘤反应率限制了PD-1/PD-L1抑制剂的临床应用。V域免疫球蛋白T细胞活化抑制因子(VISTA)是一种新型检查点调节因子,在增强抗肿瘤免疫力方面与PD-1/PD-L1具有潜在的协同作用。在此,我们报告了苯并[d]恶唑 B3 的发现,它是针对 PD-1/PD-L1 和 VISTA 的新型双重小分子抑制剂,具有很高的 PD-1/PD-L1 抑制活性和 VISTA 结合亲和力。B3 能解除 PD-L1 和 VISTA 对 T 细胞的免疫抑制,有效激活抗肿瘤免疫。此外,B3 还能诱导肿瘤细胞中 PD-L1 和 VISTA 的降解。此外,B3 在 CT26 小鼠模型中显示出显著的体内抗肿瘤疗效。我们的研究结果发现,B3是一种很有前景的PD-1/PD-L1和VISTA双重抑制剂,为克服目前抗PD-1/PD-L1疗法的局限性提供了一种新的治疗策略。
{"title":"Discovery of Benzo[d]oxazoles as Novel Dual Small-Molecule Inhibitors Targeting PD-1/PD-L1 and VISTA Pathway","authors":"Kaizhen Wang, Shi Cai, Yao Cheng, Zhihao Qi, Xiang Ni, Kuojun Zhang, Yibei Xiao, Xiangyu Zhang, Tianyu Wang","doi":"10.1021/acs.jmedchem.4c01899","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01899","url":null,"abstract":"The blockers of programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) pathway have achieved great clinical success. However, the limited efficacy and low tumor response rate of anti-PD-1/PD-L1 monotherapy limit the clinical application of PD-1/PD-L1 inhibitors. V-domain immunoglobulin suppressor of T-cell activation (VISTA), a novel checkpoint regulator, exhibits potential synergy with PD-1/PD-L1 in enhancing antitumor immunity. Herein, we report the discovery of benzo[<i>d</i>]oxazole <b>B3</b> as novel dual small-molecule inhibitors targeting PD-1/PD-L1 and VISTA with high PD-1/PD-L1 inhibitory activity and VISTA binding affinity. <b>B3</b> rescues the immunosuppression of T-cells mediated by PD-L1 and VISTA and activates antitumor immunity effectively. Moreover, <b>B3</b> could induce degradation of PD-L1 and VISTA in tumor cell. Furthermore, <b>B3</b> displays significant <i>in vivo</i> antitumor efficacy in a CT26 mouse model. Our results discover <b>B3</b> as a promising dual PD-1/PD-L1 and VISTA inhibitor, providing a novel therapeutic strategy to overcome the limitations of current anti-PD-1/PD-L1 therapy.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142398603","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery of ZLC491 as a Potent, Selective, and Orally Bioavailable CDK12/13 PROTAC Degrader 发现 ZLC491 是一种强效、选择性和可口服的 CDK12/13 PROTAC 降解剂
IF 7.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-10 DOI: 10.1021/acs.jmedchem.4c01596
Licheng Zhou, Kaijie Zhou, Yu Chang, Jianzhang Yang, Bohai Fan, Yuhan Su, Zilu Li, Rahul Mannan, Somnath Mahapatra, Ming Ding, Fengtao Zhou, Weixue Huang, Xiaomei Ren, Jian Xu, George Xiaoju Wang, Jinwei Zhang, Zhen Wang, Arul M. Chinnaiyan, Ke Ding
Selective degradation of cyclin-dependent kinases 12 and 13 (CDK12/13) emerges as a new potential therapeutic approach for triple-negative breast cancer (TNBC) and other human cancers. While several proteolysis-targeting chimera (PROTAC) degraders of CDK12/13 were reported, none are orally bioavailable. Here, we report the discovery of ZLC491 as a potent, selective, and orally bioavailable CDK12/13 PROTAC degrader. The compound effectively degraded CDK12 and CDK13 with DC50 values of 32 and 28 nM, respectively, in TNBC MDA-MB-231 cells. Global proteomic assessment and mechanistic studies revealed that ZLC491 selectively induced CDK12/13 degradation in a cereblon- and proteasome-dependent manner. Furthermore, the molecule efficiently suppressed transcription and expression of long genes, predominantly a subset of genes associated with DNA damage response, and significantly inhibited proliferation of multiple TNBC cell lines. Importantly, ZLC491 achieved an oral bioavailability of 46.8% in rats and demonstrated potent in vivo degradative effects on CDK12/13 in an MDA-MB-231 xenografted mouse model.
选择性降解细胞周期蛋白依赖性激酶 12 和 13(CDK12/13)是治疗三阴性乳腺癌(TNBC)和其他人类癌症的一种新的潜在方法。虽然有报道称CDK12/13的几种蛋白分解靶向嵌合体(PROTAC)降解剂,但没有一种可口服。在此,我们报告了 ZLC491 作为一种强效、选择性和口服生物可用性 CDK12/13 PROTAC 降解剂的发现。该化合物能有效降解 CDK12 和 CDK13,在 TNBC MDA-MB-231 细胞中的 DC50 值分别为 32 和 28 nM。全局蛋白质组学评估和机理研究显示,ZLC491以脑隆和蛋白酶体依赖的方式选择性地诱导CDK12/13降解。此外,该分子还能有效抑制长基因(主要是与DNA损伤反应相关的基因子集)的转录和表达,并显著抑制多种TNBC细胞系的增殖。重要的是,ZLC491 在大鼠体内的口服生物利用度达到 46.8%,并在 MDA-MB-231 异种移植小鼠模型中显示出对 CDK12/13 的强效体内降解作用。
{"title":"Discovery of ZLC491 as a Potent, Selective, and Orally Bioavailable CDK12/13 PROTAC Degrader","authors":"Licheng Zhou, Kaijie Zhou, Yu Chang, Jianzhang Yang, Bohai Fan, Yuhan Su, Zilu Li, Rahul Mannan, Somnath Mahapatra, Ming Ding, Fengtao Zhou, Weixue Huang, Xiaomei Ren, Jian Xu, George Xiaoju Wang, Jinwei Zhang, Zhen Wang, Arul M. Chinnaiyan, Ke Ding","doi":"10.1021/acs.jmedchem.4c01596","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01596","url":null,"abstract":"Selective degradation of cyclin-dependent kinases 12 and 13 (CDK12/13) emerges as a new potential therapeutic approach for triple-negative breast cancer (TNBC) and other human cancers. While several proteolysis-targeting chimera (PROTAC) degraders of CDK12/13 were reported, none are orally bioavailable. Here, we report the discovery of <b>ZLC491</b> as a potent, selective, and orally bioavailable CDK12/13 PROTAC degrader. The compound effectively degraded CDK12 and CDK13 with DC<sub>50</sub> values of 32 and 28 nM, respectively, in TNBC MDA-MB-231 cells. Global proteomic assessment and mechanistic studies revealed that <b>ZLC491</b> selectively induced CDK12/13 degradation in a cereblon- and proteasome-dependent manner. Furthermore, the molecule efficiently suppressed transcription and expression of long genes, predominantly a subset of genes associated with DNA damage response, and significantly inhibited proliferation of multiple TNBC cell lines. Importantly, <b>ZLC491</b> achieved an oral bioavailability of 46.8% in rats and demonstrated potent <i>in vivo</i> degradative effects on CDK12/13 in an MDA-MB-231 xenografted mouse model.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142398554","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery of Potent Azetidine-Benzoxazole MerTK Inhibitors with In Vivo Target Engagement. 发现体内靶标参与的强效氮杂环丁烷-苯并恶唑 MerTK 抑制剂
IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-10 Epub Date: 2024-09-30 DOI: 10.1021/acs.jmedchem.4c01451
Robin R Frey, Navendu Jana, Jacob V Gorman, Jin Wang, Heath A Smith, Kenneth D Bromberg, Ashish Thakur, Stella Z Doktor, Anura S Indulkar, Clarissa G Jakob, Anup K Upadhyay, Wei Qiu, Vlasios Manaves, Frank Gambino, Stephen A Valentino, Debra Montgomery, Yebin Zhou, Tao Li, Fritz G Buchanan, Debra C Ferguson, Matthew D Kurnick, Nicolas Kapecki, Albert Lai, Michael R Michaelides, Thomas D Penning

Inhibition of the receptor tyrosine kinase MerTK by small molecules has the potential to augment the immune response to tumors. Potent, selective inhibitors with high levels of in vivo target engagement are needed to fully evaluate the potential use of MerTK inhibitors as cancer therapeutics. We report the discovery and optimization of a series of pyrazinamide-based type 1.5 MerTK inhibitors bearing an azetidine-benzoxazole substituent. Compound 31 potently engages the target in vivo and demonstrates single agent activity in the immune-driven MC-38 murine syngeneic tumor model.

小分子抑制受体酪氨酸激酶 MerTK 有可能增强对肿瘤的免疫反应。要全面评估 MerTK 抑制剂作为癌症疗法的潜在用途,需要具有体内靶点高度参与性的强效选择性抑制剂。我们报告了一系列基于吡嗪酰胺的 1.5 型 MerTK 抑制剂的发现和优化,这些抑制剂带有氮杂环丁烷-苯并恶唑取代基。化合物 31 能在体内有效地与靶点结合,并在免疫驱动的 MC-38 小鼠合成肿瘤模型中显示出单药活性。
{"title":"Discovery of Potent Azetidine-Benzoxazole MerTK Inhibitors with <i>In Vivo</i> Target Engagement.","authors":"Robin R Frey, Navendu Jana, Jacob V Gorman, Jin Wang, Heath A Smith, Kenneth D Bromberg, Ashish Thakur, Stella Z Doktor, Anura S Indulkar, Clarissa G Jakob, Anup K Upadhyay, Wei Qiu, Vlasios Manaves, Frank Gambino, Stephen A Valentino, Debra Montgomery, Yebin Zhou, Tao Li, Fritz G Buchanan, Debra C Ferguson, Matthew D Kurnick, Nicolas Kapecki, Albert Lai, Michael R Michaelides, Thomas D Penning","doi":"10.1021/acs.jmedchem.4c01451","DOIUrl":"10.1021/acs.jmedchem.4c01451","url":null,"abstract":"<p><p>Inhibition of the receptor tyrosine kinase MerTK by small molecules has the potential to augment the immune response to tumors. Potent, selective inhibitors with high levels of <i>in vivo</i> target engagement are needed to fully evaluate the potential use of MerTK inhibitors as cancer therapeutics. We report the discovery and optimization of a series of pyrazinamide-based type 1.5 MerTK inhibitors bearing an azetidine-benzoxazole substituent. Compound <b>31</b> potently engages the target <i>in vivo</i> and demonstrates single agent activity in the immune-driven MC-38 murine syngeneic tumor model.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337353","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hydrazide-Based Class I Selective HDAC Inhibitors Completely Reverse Chemoresistance Synergistically in Platinum-Resistant Solid Cancer Cells. 基于酰肼的 I 类选择性 HDAC 抑制剂可协同完全逆转铂耐药性实体癌细胞的化疗耐药性。
IF 6.8 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-10 Epub Date: 2024-10-02 DOI: 10.1021/acs.jmedchem.4c01817
Fabian B Kraft, Lukas Biermann, Linda Schäker-Hübner, Maria Hanl, Alexandra Hamacher, Matthias U Kassack, Finn K Hansen

In this work, we have synthesized a set of peptoid-based histone deacetylase inhibitors (HDACi) with a substituted hydrazide moiety as zinc-binding group. Subsequently, all compounds were evaluated in biochemical HDAC inhibition assays and for their antiproliferative activity against native and cisplatin-resistant cancer cell lines. The hydrazide derivatives with a propyl or butyl substituent (compounds 5 and 6) emerged as the most potent class I HDAC selective inhibitors (HDAC1-3). Further, compounds 5 and 6 outperformed entinostat in cytotoxicity assays and were able to reverse chemoresistance in cisplatin-resistant A2780 (ovarian) and Cal27 (head-neck) cancer cell lines. Moreover, the hydrazide derivatives 5 and 6 showed strong synergism with cisplatin (combination indices <0.2), again outperforming entinostat, and increased DNA damage, p21, and pro-apoptotic BIM expression, leading to caspase-mediated apoptosis and cell death. Thus, compounds 5 and 6 represent promising lead structures for developing new HDACi capable of reversing chemoresistance in cisplatin resistant cancer cells.

在这项工作中,我们合成了一组以取代的酰肼分子为锌结合基团的肽类组蛋白去乙酰化酶抑制剂(HDACi)。随后,我们在生化 HDAC 抑制实验中对所有化合物进行了评估,并评估了它们对本地和顺铂抗性癌细胞株的抗增殖活性。具有丙基或丁基取代基的酰肼衍生物(化合物 5 和 6)成为最有效的 I 类 HDAC 选择性抑制剂(HDAC1-3)。此外,化合物 5 和 6 在细胞毒性实验中的表现优于恩替诺司他,并能逆转顺铂耐药的 A2780(卵巢癌)和 Cal27(头颈癌)细胞系的化疗耐药性。此外,酰肼衍生物 5 和 6 与顺铂表现出很强的协同作用(组合指数 5 和 6 代表了有望开发出能逆转顺铂耐药癌细胞化疗抗性的新型 HDACi 的先导结构)。
{"title":"Hydrazide-Based Class I Selective HDAC Inhibitors Completely Reverse Chemoresistance Synergistically in Platinum-Resistant Solid Cancer Cells.","authors":"Fabian B Kraft, Lukas Biermann, Linda Schäker-Hübner, Maria Hanl, Alexandra Hamacher, Matthias U Kassack, Finn K Hansen","doi":"10.1021/acs.jmedchem.4c01817","DOIUrl":"10.1021/acs.jmedchem.4c01817","url":null,"abstract":"<p><p>In this work, we have synthesized a set of peptoid-based histone deacetylase inhibitors (HDACi) with a substituted hydrazide moiety as zinc-binding group. Subsequently, all compounds were evaluated in biochemical HDAC inhibition assays and for their antiproliferative activity against native and cisplatin-resistant cancer cell lines. The hydrazide derivatives with a propyl or butyl substituent (compounds <b>5</b> and <b>6</b>) emerged as the most potent class I HDAC selective inhibitors (HDAC1-3). Further, compounds <b>5</b> and <b>6</b> outperformed entinostat in cytotoxicity assays and were able to reverse chemoresistance in cisplatin-resistant A2780 (ovarian) and Cal27 (head-neck) cancer cell lines. Moreover, the hydrazide derivatives <b>5</b> and <b>6</b> showed strong synergism with cisplatin (combination indices <0.2), again outperforming entinostat, and increased DNA damage, p21, and pro-apoptotic BIM expression, leading to caspase-mediated apoptosis and cell death. Thus, compounds <b>5</b> and <b>6</b> represent promising lead structures for developing new HDACi capable of reversing chemoresistance in cisplatin resistant cancer cells.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142360752","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Discovery of Propionic Acid Derivatives with a 5-THIQ Core as Potent and Orally Bioavailable Keap1–Nrf2 Protein–Protein Interaction Inhibitors for Acute Kidney Injury 发现以 5-THIQ 为核心的丙酸衍生物,作为强效口服生物活性 Keap1-Nrf2 蛋白-蛋白相互作用抑制剂治疗急性肾损伤
IF 7.3 1区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-10-10 DOI: 10.1021/acs.jmedchem.4c01687
Zeyu Shi, Yong Zhang, Xinyu Wang, Jingshu Tang, Yuying Kang, Jiahuan Hu, Li Li, Beibei Yang, Si Chen, Qiong Xiao, Jiaqi Lan, Jinping Hu, Ying Peng, Dali Yin
Keap1 plays a crucial role in regulating the Nrf2-mediated cytoprotective response and is increasingly targeted for oxidative stress-related diseases. Using small molecules to disrupt the Keap1–Nrf2 protein–protein interaction (PPI) has emerged as a new strategy for developing Nrf2 activators. Through extensive structure–activity relationship studies, we identified compound 56, which features a unique 5-tetrahydroisoquinoline scaffold and acts as a potent inhibitor of the Keap1–Nrf2 PPI. Compound 56 exhibited significant inhibitory activity (IC50 = 16.0 nM) and tight Keap1 binding affinity (Kd = 3.07 nM), along with acceptable oral bioavailability (F = 20%). Notably, 56 enhanced antioxidant defenses in HK-2 renal tubular epithelial cells and significantly reduced plasma creatinine and blood urea nitrogen levels in acute kidney injury (AKI) mice. These findings collectively position compound 56 as a promising candidate for the treatment of AKI.
Keap1 在调节 Nrf2 介导的细胞保护反应中发挥着至关重要的作用,并日益成为氧化应激相关疾病的靶向药物。利用小分子破坏 Keap1-Nrf2 蛋白-蛋白相互作用(PPI)已成为开发 Nrf2 激活剂的一种新策略。通过广泛的结构-活性关系研究,我们发现了化合物 56,它具有独特的 5-四氢异喹啉支架,是 Keap1-Nrf2 PPI 的强效抑制剂。化合物 56 具有明显的抑制活性(IC50 = 16.0 nM)和与 Keap1 紧密结合的亲和力(Kd = 3.07 nM),同时具有可接受的口服生物利用度(F = 20%)。值得注意的是,56 能增强 HK-2 肾小管上皮细胞的抗氧化防御能力,并显著降低急性肾损伤(AKI)小鼠的血浆肌酐和血尿素氮水平。这些发现共同将 56 号化合物定位为治疗 AKI 的有望候选药物。
{"title":"Discovery of Propionic Acid Derivatives with a 5-THIQ Core as Potent and Orally Bioavailable Keap1–Nrf2 Protein–Protein Interaction Inhibitors for Acute Kidney Injury","authors":"Zeyu Shi, Yong Zhang, Xinyu Wang, Jingshu Tang, Yuying Kang, Jiahuan Hu, Li Li, Beibei Yang, Si Chen, Qiong Xiao, Jiaqi Lan, Jinping Hu, Ying Peng, Dali Yin","doi":"10.1021/acs.jmedchem.4c01687","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01687","url":null,"abstract":"Keap1 plays a crucial role in regulating the Nrf2-mediated cytoprotective response and is increasingly targeted for oxidative stress-related diseases. Using small molecules to disrupt the Keap1–Nrf2 protein–protein interaction (PPI) has emerged as a new strategy for developing Nrf2 activators. Through extensive structure–activity relationship studies, we identified compound <b>56</b>, which features a unique 5-tetrahydroisoquinoline scaffold and acts as a potent inhibitor of the Keap1–Nrf2 PPI. Compound <b>56</b> exhibited significant inhibitory activity (IC<sub>50</sub> = 16.0 nM) and tight Keap1 binding affinity (<i>K</i><sub>d</sub> = 3.07 nM), along with acceptable oral bioavailability (<i>F</i> = 20%). Notably, <b>56</b> enhanced antioxidant defenses in HK-2 renal tubular epithelial cells and significantly reduced plasma creatinine and blood urea nitrogen levels in acute kidney injury (AKI) mice. These findings collectively position compound <b>56</b> as a promising candidate for the treatment of AKI.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142398548","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Medicinal Chemistry
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