Journal of Medicinal Chemistry最新文献

筛选
英文 中文
Discovery of ZG-2305, an Orally Bioavailable Factor Inhibiting HIF Inhibitor for the Treatment of Obesity and Fatty Liver Disease 发现用于治疗肥胖症和脂肪肝的口服生物活性因子抑制剂 ZG-2305
IF 7.3 1区 医学
Journal of Medicinal Chemistry Pub Date : 2024-10-21 DOI: 10.1021/acs.jmedchem.4c01698
Yue Wu, Zewei Zhang, Haiping Cai, Weiqing Zhang, Linjian Zhang, Zhihong Li, Le Yang, Yafen Chen, Thomas P. Corner, Zhe Song, Jie Yue, Fulai Yang, Xiang Li, Christopher J. Schofield, Xiaojin Zhang
{"title":"Discovery of ZG-2305, an Orally Bioavailable Factor Inhibiting HIF Inhibitor for the Treatment of Obesity and Fatty Liver Disease","authors":"Yue Wu, Zewei Zhang, Haiping Cai, Weiqing Zhang, Linjian Zhang, Zhihong Li, Le Yang, Yafen Chen, Thomas P. Corner, Zhe Song, Jie Yue, Fulai Yang, Xiang Li, Christopher J. Schofield, Xiaojin Zhang","doi":"10.1021/acs.jmedchem.4c01698","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01698","url":null,"abstract":"Genetic loss of the 2-oxoglutarate oxygenase factor inhibiting hypoxia-inducible factor (FIH) enhances both glycolysis and aerobic metabolism. FIH is thus a potential target for adiposity control and improving hepatic steatosis. We describe development of a series of novel, potent, and selective FIH inhibitors that occupy both the FIH catalytic site and a recently defined tyrosine conformational-flip pocket. <b>ZG-2305</b>, with a <i>K</i><sub>i</sub> of 79.6 nM for FIH, manifests 38-fold selectivity over the hypoxia-inducible factor (HIF) prolyl hydroxylase PHD2. Oral administration of <b>ZG-2305</b> in the western-diet induced obesity mouse model results in improved lipid accumulation and recovery from abnormal body weight/hepatic steatosis. Amelioration of nonalcoholic steatohepatitis (NASH) related pathological phenotypes in the HF-CDAA-diet induced NASH mouse model was observed. Preliminary preclinical studies indicate <b>ZG-2305</b> has good pharmacokinetic properties and an acceptable safety profile. The results imply <b>ZG-2305</b> is a promising candidate for treatment of obesity and fatty liver disease.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142452329","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 Arene Ruthenium(II) Complexes as Potential VEGF Inhibitors for Glioblastoma Metastasis Suppression 发现作为潜在血管内皮生长因子抑制剂的芘钌(II)配合物以抑制胶质母细胞瘤转移
IF 7.3 1区 医学
Journal of Medicinal Chemistry Pub Date : 2024-10-21 DOI: 10.1021/acs.jmedchem.4c00797
Chanling Yuan, Chunguang Zhu, Qingshuang Lv, Jiahui Shi, Jiacheng Wang, Shiqi Gao, Jiayi Qian, Yanhua Chen, Qiong Wu, Wenjie Mei
{"title":"Discovery of Arene Ruthenium(II) Complexes as Potential VEGF Inhibitors for Glioblastoma Metastasis Suppression","authors":"Chanling Yuan, Chunguang Zhu, Qingshuang Lv, Jiahui Shi, Jiacheng Wang, Shiqi Gao, Jiayi Qian, Yanhua Chen, Qiong Wu, Wenjie Mei","doi":"10.1021/acs.jmedchem.4c00797","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c00797","url":null,"abstract":"Developing drugs for treating glioblastoma has been a significant challenge. Herein, a series of arene ruthenium(II) complexes have been synthesized and investigated as potential candidates to suppress the proliferation and metastasis of glioblastoma. It is found that para-substituent-modified molecules, especially <b>6</b>, exhibit higher antitumor activity than ortho-substituents. Further studies show that <b>6</b> can trigger tumor cell autophagy by regulating the PI3K/AKT/mTOR pathway. Moreover, it is also found that <b>6</b> can induce DNA damage in glioblastoma cells through binding and stabilizing VEGF G-quadruplex DNA. Furthermore, it is confirmed that <b>6</b> can inhibit the proliferation and metastasis of U87-MG glioblastoma cell in situ xenograft in the zebrafish model. Hence, arene ruthenium(II) complexes can be developed as promising therapeutic agents for glioblastoma treatment in the future.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142452218","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
Structure-Affinity-Pharmacokinetics Relationships of Novel 18F-Labeled 1,4-Diazepane Derivatives for Orexin 1 Receptor Imaging 用于奥列克辛 1 受体成像的新型 18F 标记 1,4-二氮杂环庚烷衍生物的结构-亲和力-药代动力学关系
IF 7.3 1区 医学
Journal of Medicinal Chemistry Pub Date : 2024-10-21 DOI: 10.1021/acs.jmedchem.4c01090
Yui Ishizaka, Hiroyuki Watanabe, Masahiro Ono
{"title":"Structure-Affinity-Pharmacokinetics Relationships of Novel 18F-Labeled 1,4-Diazepane Derivatives for Orexin 1 Receptor Imaging","authors":"Yui Ishizaka, Hiroyuki Watanabe, Masahiro Ono","doi":"10.1021/acs.jmedchem.4c01090","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01090","url":null,"abstract":"The orexin 1 receptor (OX1R) has been suggested to be involved in the reward and autonomic nervous systems. Positron emission tomography (PET) of OX1R contributes to elucidating its role and developing new drugs. However, there are no useful PET probes for in vivo imaging of OX1R. Here, we newly designed and synthesized <sup>18</sup>F-labeled 1,4-diazepane derivatives and evaluated their utilities as OX1R PET probes. In particular, BTF showed high and selective binding affinity for OX1R. In a biodistribution study using normal mice, [<sup>18</sup>F]BTF exhibited brain uptake, and radioactivity in the brain was significantly decreased by preinjection of unlabeled BTF. In a PET/CT study, it was suggested that [<sup>18</sup>F]BTF has the potential to visualize high-expression regions of OX1R in the normal mouse brain. Collectively, [<sup>18</sup>F]BTF has the fundamental features of an OX1R PET probe, and further studies may lead to the development of more useful probes.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142451894","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
Cell-Based Screen Identifies a Highly Potent and Orally Available ABCB1 Modulator for Treatment of Multidrug Resistance 基于细胞的筛选发现了一种用于治疗多药耐药性的强效口服 ABCB1 调节剂
IF 7.3 1区 医学
Journal of Medicinal Chemistry Pub Date : 2024-10-19 DOI: 10.1021/acs.jmedchem.4c01081
Shuai Wang, Sai-Qi Wang, Xiao-Bing Chen, Qian Xu, Hao Deng, Qiu-Xu Teng, Zhe-Sheng Chen, Xuyao Zhang, Fen-Er Chen
{"title":"Cell-Based Screen Identifies a Highly Potent and Orally Available ABCB1 Modulator for Treatment of Multidrug Resistance","authors":"Shuai Wang, Sai-Qi Wang, Xiao-Bing Chen, Qian Xu, Hao Deng, Qiu-Xu Teng, Zhe-Sheng Chen, Xuyao Zhang, Fen-Er Chen","doi":"10.1021/acs.jmedchem.4c01081","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01081","url":null,"abstract":"Targeting ABCB1 is a promising strategy in combating multidrug resistance. Our cell-based phenotypic screening led to the discovery of novel triazolo[1,5-<i>a</i>]pyrimidone-based ABCB1 modulators. Notably, <b>WS-917</b> was identified as a significant contributor to heightened sensitization of human colorectal adenocarcinoma cells (SW620/Ad300) to paclitaxel (IC<sub>50</sub> = 5 nM). Mechanistic elucidation revealed that this compound substantially augmented intracellular paclitaxel and [<sup>3</sup>H]-paclitaxel, concurrently mitigating the efflux of [<sup>3</sup>H]-paclitaxel in SW620/Ad300 through the inhibition of ABCB1 efflux. The cellular thermal shift assay underscored its ability to stabilize ABCB1 through direct binding. Additionally, <b>WS-917</b> induced stimulation of ABCB1 ATPase activity while exhibiting negligible inhibitory effect against CYP3A4. Remarkable was its capacity to enhance the sensitivity of SW620/Ad300 to paclitaxel, as well as the sensitivity of CT26/TAXOL to paclitaxel and PD-L1 inhibitor (Atezolizumab) in vivo, all achieved without inducing observable toxicity. The discovery of <b>WS-917</b> holds promise for the development of more potent ABCB1 modulators.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142449732","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
Covalent Inhibitors of S100A4 Block the Formation of a Pro-Metastasis Non-Muscle Myosin 2A Complex S100A4的共价抑制剂能阻止形成促进转移的非肌球蛋白2A复合物
IF 7.3 1区 医学
Journal of Medicinal Chemistry Pub Date : 2024-10-19 DOI: 10.1021/acs.jmedchem.4c01320
Charline Giroud, Tamas Szommer, Carmen Coxon, Octovia Monteiro, Thomas Grimes, Tryfon Zarganes-Tzitzikas, Thomas Christott, James Bennett, Karly Buchan, Paul E. Brennan, Oleg Fedorov
{"title":"Covalent Inhibitors of S100A4 Block the Formation of a Pro-Metastasis Non-Muscle Myosin 2A Complex","authors":"Charline Giroud, Tamas Szommer, Carmen Coxon, Octovia Monteiro, Thomas Grimes, Tryfon Zarganes-Tzitzikas, Thomas Christott, James Bennett, Karly Buchan, Paul E. Brennan, Oleg Fedorov","doi":"10.1021/acs.jmedchem.4c01320","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01320","url":null,"abstract":"The S100 protein family functions as protein–protein interaction adaptors regulated by Ca<sup>2+</sup> binding. Formation of various S100 complexes plays a central role in cell functions, from calcium homeostasis to cell signaling, and is implicated in cell growth, migration, and tumorigenesis. We established a suite of biochemical and cellular assays for small molecule screening based on known S100 protein–protein interactions. From 25 human S100 proteins, we focused our attention on S100A4 because of its well-established role in cancer progression and metastasizes by interacting with nonmuscle myosin II (NMII). We identified several potent and selective inhibitors of this interaction and established the covalent nature of binding, confirmed by mass spectrometry and crystal structures. <b>5b</b> showed on-target activity in cells and inhibition of cancer cell migration. The identified S100A4 inhibitors can serve as a basis for the discovery of new cancer drugs operating via a novel mode of action.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142450099","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
Optimization of B-Ring-Functionalized Antimalarial Tambjamines and Prodiginines 优化 B 环功能化抗疟坦贾明和丙二炔类化合物
IF 7.3 1区 医学
Journal of Medicinal Chemistry Pub Date : 2024-10-19 DOI: 10.1021/acs.jmedchem.4c02093
Amrendra Kumar, Sivanna Chithanna, Yuexin Li, Xiaowei Zhang, Rozalia A. Dodean, Diana Caridha, Michael S. Madejczyk, Patricia J. Lee, Xiannu Jin, Ravi Chetree, Cameron Blount, William E. Dennis, Jesse DeLuca, Chau Vuong, Kristina Pannone, Hieu T. Dinh, Susan Leed, Alison Roth, Kevin A. Reynolds, Jane X. Kelly, Papireddy Kancharla
{"title":"Optimization of B-Ring-Functionalized Antimalarial Tambjamines and Prodiginines","authors":"Amrendra Kumar, Sivanna Chithanna, Yuexin Li, Xiaowei Zhang, Rozalia A. Dodean, Diana Caridha, Michael S. Madejczyk, Patricia J. Lee, Xiannu Jin, Ravi Chetree, Cameron Blount, William E. Dennis, Jesse DeLuca, Chau Vuong, Kristina Pannone, Hieu T. Dinh, Susan Leed, Alison Roth, Kevin A. Reynolds, Jane X. Kelly, Papireddy Kancharla","doi":"10.1021/acs.jmedchem.4c02093","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c02093","url":null,"abstract":"Malaria has been a deadly enemy of mankind throughout history, affecting over 200 million people annually, along with approximately half a million deaths. Resistance to current therapies is of great concern, and there is a dire need for novel and well-tolerated antimalarials that operate by clinically unexploited mechanisms. We have previously reported that both tambjamines and prodiginines are highly potent novel antiplasmodial agents, but they required rigor optimizations to enhance the oral efficacy, safety, and physicochemical properties. Here, we launched a comprehensive structure–activity relationship study for B-ring-functionalized tambjamines and prodiginines with 54 novel analogues systematically designed and synthesized. A number of compounds exhibited remarkable antiplasmodial activities against asexual erythrocytic <i>Plasmodium</i> parasites, with improved safety and metabolic profiles. Notably, several prodiginines cured erythrocytic <i>Plasmodium yoelii</i> infections after oral 25 mg/kg × 4 days in a murine model and provided partial protection against liver stage <i>Plasmodium berghei</i> sporozoite-induced infection in mice.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142450010","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
Correction to “Discovery of Novel Histone Deacetylase 6 (HDAC6) Inhibitors with Enhanced Antitumor Immunity of Anti-PD-L1 Immunotherapy in Melanoma” 对 "发现新型组蛋白去乙酰化酶 6 (HDAC6) 抑制剂,增强黑色素瘤中抗 PD-L1 免疫疗法的抗肿瘤免疫力 "的更正
IF 7.3 1区 医学
Journal of Medicinal Chemistry Pub Date : 2024-10-19 DOI: 10.1021/acs.jmedchem.4c02486
Xiaopeng Peng, Ling Li, Jingxuan Chen, Yichang Ren, Jin Liu, Ziwen Yu, Hao Cao, Jianjun Chen
{"title":"Correction to “Discovery of Novel Histone Deacetylase 6 (HDAC6) Inhibitors with Enhanced Antitumor Immunity of Anti-PD-L1 Immunotherapy in Melanoma”","authors":"Xiaopeng Peng, Ling Li, Jingxuan Chen, Yichang Ren, Jin Liu, Ziwen Yu, Hao Cao, Jianjun Chen","doi":"10.1021/acs.jmedchem.4c02486","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c02486","url":null,"abstract":"In our study to investigate the impact of two newly synthesized compounds (XP5 and XP19) alongside a positive control drug (CAY10603) on the expression levels of various target proteins─specifically, AC-α-Tubulin, α-Tubulin, SMC3, Ac-H3, H3, Ac-SMC3, and apoptosis-related antibodies (Ac-Ku70, Ku70, Bcl-2, Bax)─we conducted a series of experiments on different tumor cell lines, including Jurkat-T cells and B16-F10 cells. This comprehensive analysis yielded a substantial amount of experimental data and protein bands. Regrettably, during the assembly of Figure 7 and Figure 8 in the original manuscript, we inadvertently used a wrong image of Ac-Ku70 and Ku70 in Figure 7A and of Ac-SMC3, SMC3, and Ac-H3 in Figure 8E,F. Upon recent and careful review of the original manuscript, we identified this error and have replaced the wrong image with the correct one in the corrected Figure 7 and Figure 8. This corrected result still demonstrates that XP5 and XP19 can upregulate the expression levels of acetylated Ku70 significantly and dose-dependently but showed no effect on the Ku70 levels and had little effect on the levels of acetyl-SMC3 and acetyl-H3, and these corrections do not change the scientific conclusions of the article in any way. Figure 7. Analysis of acetylated Ku70, Ku70, Bcl-2, and Bax by Western blot. Jurkat T cells were treated with dimethyl sulfoxide (DMSO) or XP5, and XP19 for 6 h. The levels of acetylated Ku70, Ku70, Bcl-2, and Bax were examined by Western blot (A). Quantitative analysis of the levels of acetylated Ku70 (B), Bcl-2 (C), and Bax (D). All data are representative of three independent experiments and shown as mean ± SD. ***&lt;i&gt;p&lt;/i&gt; &lt; 0.001, **&lt;i&gt;p&lt;/i&gt; &lt; 0.01, *&lt;i&gt;p&lt;/i&gt; &lt; 0.05 compared with the control group, &lt;sup&gt;###&lt;/sup&gt;&lt;i&gt;p&lt;/i&gt; &lt; 0.001. One-way ANOVA for the above analysis, Dunnett test. Figure 8. Analysis of acetylated α-tubulin/acetylated-SMC3 and acetylated-H3 by Western blot. The levels of acetyl-α-tubulin (Ac-α-Tub) and α-tubulin in B16-F10 cells (A) and Jurkat T cells (B) treated with DMSO or XP5, XP19, and CAY10603 for 6 h. Quantitative analysis of the level of acetyl-α-tubulin (Ac-α-Tub) by Western blotting assay obtained in B16-F10 cells (C) and Jurkat T cells (D). The levels of acetylated-SMC3, total SMC3, acetylated-H3, and total H3 in B16-F10 cells (E) and Jurkat T cells (F) treated with DMSO or XP5, XP19, and CAY10603 for 6 h. All data are representative of three independent experiments and shown as mean ± SD. ***&lt;i&gt;p&lt;/i&gt; &lt; 0.001, **&lt;i&gt;p&lt;/i&gt; &lt; 0.01 compared with the control group, &lt;sup&gt;###&lt;/sup&gt;&lt;i&gt;p&lt;/i&gt; &lt; 0.001, &lt;sup&gt;&amp;&lt;/sup&gt;&lt;i&gt;p&lt;/i&gt; &lt; 0.05, &lt;sup&gt;&amp;&amp;&lt;/sup&gt;&lt;i&gt;p&lt;/i&gt; &lt; 0.01. One-way ANOVA for the above analysis, Dunnett test. We have now corrected the original Figure 7 and Figure 8, and all authors have agreed to the changes. We apologize for any confusion this may have caused and appreciate your understanding as we make these necessary corrections to ensure the integrity of our","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142450012","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 Bis-imidazolecarboxamide Derivatives as Novel, Potent, and Selective TNIK Inhibitors for the Treatment of Idiopathic Pulmonary Fibrosis 发现双咪唑甲酰胺衍生物作为治疗特发性肺纤维化的新型、强效和选择性 TNIK 抑制剂
IF 7.3 1区 医学
Journal of Medicinal Chemistry Pub Date : 2024-10-18 DOI: 10.1021/acs.jmedchem.4c01580
Vladimir Aladinskiy, Chris Kruse, Luoheng Qin, Eugene Babin, Yaya Fan, Georgiy Andreev, Heng Zhao, Yanyun Fu, Man Zhang, Yan Ivanenkov, Alex Aliper, Alex Zhavoronkov, Feng Ren
{"title":"Discovery of Bis-imidazolecarboxamide Derivatives as Novel, Potent, and Selective TNIK Inhibitors for the Treatment of Idiopathic Pulmonary Fibrosis","authors":"Vladimir Aladinskiy, Chris Kruse, Luoheng Qin, Eugene Babin, Yaya Fan, Georgiy Andreev, Heng Zhao, Yanyun Fu, Man Zhang, Yan Ivanenkov, Alex Aliper, Alex Zhavoronkov, Feng Ren","doi":"10.1021/acs.jmedchem.4c01580","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01580","url":null,"abstract":"Traf2- and Nck-interacting kinase (TNIK) has been identified as a promising therapeutic target for the treatment of fibrosis-driven diseases. Utilizing a structure-based drug design workflow, we developed a series of potent TNIK inhibitors that modulate the conformation of the gatekeeper Met105 side chain and access the TNIK back pocket. The lead optimization efforts culminated in the discovery of the recently reported compound <b>4</b> (INS018_055), a novel TNIK inhibitor. This molecule demonstrated excellent activity in both enzymatic and cell-based assays, along with high selectivity in a kinome panel. Further, <i>in vitro</i> and <i>in vivo</i> preclinical studies revealed favorable <i>in vitro</i> and <i>in vivo</i> DMPK properties. Results from multiple cell-based and animal models proved that compound <b>4</b> exhibits considerable antifibrotic and anti-inflammatory efficacy. Currently, phase II clinical trials of compound <b>4</b> are underway for the treatment of idiopathic pulmonary fibrosis (IPF).","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142448583","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
Advancing Cancer Immunotherapy through Engineering New PD-L1 Degraders: A Comprehensive Study from Small Molecules to PD-L1-Specific Peptide–Drug Conjugates 通过设计新的 PD-L1 降解剂推进癌症免疫疗法:从小分子到 PD-L1 特异性肽-药物共轭物的综合研究
IF 7.3 1区 医学
Journal of Medicinal Chemistry Pub Date : 2024-10-18 DOI: 10.1021/acs.jmedchem.4c01652
Zekun Zeng, Zhiwei Yang, Chenghao Li, Shujing Liu, Wei Wei, Ye Zhou, Simeng Wang, Mengjun Sui, Mengdan Li, Shumei Lin, Yangyang Cheng, Peng Hou
{"title":"Advancing Cancer Immunotherapy through Engineering New PD-L1 Degraders: A Comprehensive Study from Small Molecules to PD-L1-Specific Peptide–Drug Conjugates","authors":"Zekun Zeng, Zhiwei Yang, Chenghao Li, Shujing Liu, Wei Wei, Ye Zhou, Simeng Wang, Mengjun Sui, Mengdan Li, Shumei Lin, Yangyang Cheng, Peng Hou","doi":"10.1021/acs.jmedchem.4c01652","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01652","url":null,"abstract":"Despite the considerable achievements of antibodies targeting PD-1/PD-L1 in cancer immunotherapy, limitations in antitumor immune response and pharmacokinetics hinder their clinical adoption. Small molecules toward PD-L1 degradation signifies an innovative avenue to modulate PD-1/PD-L1 axis. Herein, we unveil a comprehensive engineering involving the development of new PD-L1 degraders based on the berberine (BBR) and palmatine (PMT) bioactive frameworks and explore their translational potential for cancer immunotherapy using a peptide-drug conjugate strategy. Chemical modifications at the O-9 position of PMT dramatically enhance the PD-L1 degradation capacity. Further conjugation of PMT degraders with an anti-PD-L1 peptide featuring disulfide linkers enables efficient GSH-specific prodrug activation, yielding synergistic immunotherapeutic benefits through both external PD-L1 blockade and internal PD-L1 degradation mechanisms. This work elucidates the compelling charm of the discovery and application of PD-L1 degraders, offering solutions to the challenges in advancing cancer immunotherapy in widespread clinics.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142448589","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
Correction to “The New Frontier: Merging Molecular Glue Degrader and Antibody–Drug Conjugate Modalities To Overcome Strategic Challenges” 更正 "新前沿:合并分子胶水降解剂和抗体药物共轭物模式以克服战略挑战"
IF 7.3 1区 医学
Journal of Medicinal Chemistry Pub Date : 2024-10-18 DOI: 10.1021/acs.jmedchem.4c02447
Yam B. Poudel, Ruchita R. Thakore, Eugene P. Chekler
{"title":"Correction to “The New Frontier: Merging Molecular Glue Degrader and Antibody–Drug Conjugate Modalities To Overcome Strategic Challenges”","authors":"Yam B. Poudel, Ruchita R. Thakore, Eugene P. Chekler","doi":"10.1021/acs.jmedchem.4c02447","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c02447","url":null,"abstract":"The authors regret an inadvertent error in Figure 1 where in both label 3, the “lysosome” should read “late endosome”. We apologize for the inconvenience. This article has not yet been cited by other publications.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-10-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142448590","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信