Jinlong Li, Tong Zhang, Di Wu, Chen He, Haoxiang Weng, Tiandong Zheng, Jie Liu, Hong Yao, Jichao Chen, Yansong Ren, Zheying Zhu, Jinyi Xu, Shengtao Xu
{"title":"Palladium-Mediated Bioorthogonal System for Prodrug Activation of <i>N</i>-Benzylbenzamide-Containing Tubulin Polymerization Inhibitors for the Treatment of Solid Tumors.","authors":"Jinlong Li, Tong Zhang, Di Wu, Chen He, Haoxiang Weng, Tiandong Zheng, Jie Liu, Hong Yao, Jichao Chen, Yansong Ren, Zheying Zhu, Jinyi Xu, Shengtao Xu","doi":"10.1021/acs.jmedchem.4c02419","DOIUrl":"10.1021/acs.jmedchem.4c02419","url":null,"abstract":"<p><p>Bioorthogonal cleavage reactions have been developed as an intriguing strategy to enhance the safety of chemotherapeutics. Aiming to reduce the toxicity and improve the targeted release properties of the colchicine binding site inhibitors (CBSIs) based on previous work, a series of biologically inert prodrugs were further designed and synthesized through a bioorthogonal prodrug strategy. The therapeutic effects of prodrugs could be \"turned-on\" once combined with palladium resins. Particularly, prodrug <b>2b</b> was 68.3-fold less cytotoxic compared to the parent compound, while its cytotoxicity was recovered <i>in situ</i> in the presence of palladium resins. Mechanism studies confirmed that <b>2b</b> inhibited cell growth in the same manner as CBSIs. More importantly, <i>in vivo</i> efficacy studies demonstrated the efficient activation of <b>2b</b> by palladium resins, resulting in significant inhibition of tumor growth (63.2%). These results suggest that prodrug <b>2b</b> with improved safety and targeted release property catalyzed by a Pd-mediated bioorthogonal cleavage reaction deserves further investigation.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142556627","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}
Jinbiao Liao, Jianing Liao, Minkui Zhang, Yanzhen Yu, Lvtao Cai, Kaixin Le, Weitao Fu, Yiyang Qin, Tingjun Hou, Dan Li, Rong Sheng
{"title":"Identification of Oral Bioavailable Coumarin Derivatives as Potential AR Antagonists Targeting Prostate Cancer.","authors":"Jinbiao Liao, Jianing Liao, Minkui Zhang, Yanzhen Yu, Lvtao Cai, Kaixin Le, Weitao Fu, Yiyang Qin, Tingjun Hou, Dan Li, Rong Sheng","doi":"10.1021/acs.jmedchem.4c01752","DOIUrl":"10.1021/acs.jmedchem.4c01752","url":null,"abstract":"<p><p>Androgen receptor (AR) is a crucial driver of prostate cancer (PCa), but acquired resistance to AR antagonists significantly undermines their clinical efficacy. We previously discovered coumarin derivative <b>1</b>, which is capable of disrupting AR ligand-binding domain dimers, offering the potential for overcoming resistance. However, its poor oral bioavailability limited further development. In this study, comprehensive structure optimizations led to compound <b>4a</b> (IC<sub>50</sub> = 0.051 μM), which exhibited comparable AR antagonistic activity to enzalutamide (IC<sub>50</sub> = 0.060 μM) and demonstrated excellent selectivity over other nuclear receptors in vitro. Especially, <b>4a</b> showed superior efficacy against AR<sup>F876L/T877A</sup> and AR<sup>W741C</sup> mutants compared to darolutamide and enzalutamide. Moreover, <b>4a</b> exhibited favorable pharmacokinetic profiles (<i>F</i> = 66.24%) in vivo and significant tumor growth inhibition in an LNCaP xenograft mouse model upon oral administration. These results highlight the potential of <b>4a</b> as a promising oral AR antagonist for overcoming drug resistance in PCa.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142566388","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}
Rizliya Visvanathan, Dang Truong Le, Sushil Dhital, Topul Rali, Rohan A Davis, Gary Williamson
{"title":"Inhibition of Human Salivary and Pancreatic α-Amylase by Resveratrol Oligomers.","authors":"Rizliya Visvanathan, Dang Truong Le, Sushil Dhital, Topul Rali, Rohan A Davis, Gary Williamson","doi":"10.1021/acs.jmedchem.4c01042","DOIUrl":"10.1021/acs.jmedchem.4c01042","url":null,"abstract":"<p><p>A key strategy to mitigate postprandial hyperglycemia involves inhibiting α-amylases, which commence the starch digestion process in the gut. This study examined the inhibitory effects of resveratrol and stilbenoid tetramers, vaticanol B, (-)-hopeaphenol, and vatalbinoside A on human salivary and pancreatic α-amylases experimentally and through molecular docking studies. Vaticanol B demonstrated the most potent inhibition with IC<sub>50</sub> values of 5.3 ± 0.3 μM for salivary and 6.1 ± 0.5 μM for pancreatic α-amylase (compared to acarbose with IC<sub>50</sub> values of 1.2 ± 0.1 μM and 0.5 ± 0.0 μM, respectively). Kinetic analysis suggested a competitive inhibition mode for vaticanol B. Resveratrol and vatalbinoside A were poor inhibitors of human α-amylases, while (-)-hopeaphenol exhibited moderate inhibition. Molecular docking supported the inhibition data, and several aspects of the structural configurations explained the stronger inhibition exerted by vaticanol B. Overall, vaticanol B shows promise as a natural alternative to acarbose for inhibiting α-amylase.</p>","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":6.8,"publicationDate":"2024-11-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142580832","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}
Dengqin He, Gang Li, Jia-Qiang Wu, Yan Geng, Xudong Qian, Yuanhui Liu, Yanghui Ou, Mengjie Li, Jun Wang, Wei Pan, Guoping Zhang, Dandan Chen, Jiaxin Chen, Zichen Xu, Hengming Ke, Hongliang Yao
{"title":"Design, Synthesis, and Biological Evaluation of New Selective PDE4 Inhibitors for Topical Treatment of Psoriasis","authors":"Dengqin He, Gang Li, Jia-Qiang Wu, Yan Geng, Xudong Qian, Yuanhui Liu, Yanghui Ou, Mengjie Li, Jun Wang, Wei Pan, Guoping Zhang, Dandan Chen, Jiaxin Chen, Zichen Xu, Hengming Ke, Hongliang Yao","doi":"10.1021/acs.jmedchem.4c01804","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01804","url":null,"abstract":"Psoriasis is a complex and chronic inflammatory disease. Current drugs help control the symptoms of psoriasis but make no cure, urging discovery of novel drugs. We report in this paper the discovery of new phosphodiesterase 4 (PDE4) inhibitors for treatment of psoriasis. We designed and synthesized 45 new compounds, among which <b>14h</b> exhibited IC<sub>50</sub> of 0.57 nM for PDE4D and >4100-fold selectivity over other PDE families. Compound <b>14h</b> inhibited release of inflammatory cytokines of TNF-α (IC<sub>50</sub> = 34.2 μM) and IL-6 (IC<sub>50</sub> = 40.9 μM) in Raw264.7 cells and reduced the expression of IL-1β and IL-17A in the skin of psoriasis mice. In addition, <b>14h</b> alleviated IMQ-induced psoriasis in the mouse model and reduced the erythema level, scales, and thickness of the back skin of the mice. In short, our results suggested that PDE4 inhibitor <b>14h</b> is a strong candidate for the topical treatment of psoriasis.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142601195","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}
Shiliang Hu, Linjiang Tong, Qiao Qin, Jiaxin Wen, Yan Li, Fang Feng, Kunzhong Wu, Yang Zhou, Jinsai Shang, Junjian Wang, Jinbao Liu, Hua Xie, Xiaoyun Lu
{"title":"Design, Synthesis, and Biological Evaluation of Novel Diaminopyrimidine Macrocycles as Fourth Generation Reversible EGFR Inhibitors That Overcome Clinical Resistance to Osimertinib Mediated by C797S Mutation","authors":"Shiliang Hu, Linjiang Tong, Qiao Qin, Jiaxin Wen, Yan Li, Fang Feng, Kunzhong Wu, Yang Zhou, Jinsai Shang, Junjian Wang, Jinbao Liu, Hua Xie, Xiaoyun Lu","doi":"10.1021/acs.jmedchem.4c01975","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01975","url":null,"abstract":"Overcoming clinical resistance to osimertinib mediated by the tertiary C797S mutation remains an unmet medical need. To date, there are no effective drugs that have been approved for patients who harbor EGFR<sup>T790M/C797S</sup> mutations. Herein, we applied a structure-based drug design strategy to discover a series of potent and selective diaminopyrimidine macrocycles as novel EGFR<sup>T790M/C797S</sup> inhibitors. The representative compound <b>21v</b> potently inhibited EGFR<sup>19del/T790M/C797S</sup> and EGFR<sup>L858R/T790M/C797S</sup> mutants with IC<sub>50</sub> values of 2.3 nM and 12.5 nM, respectively, and exhibited antiproliferative activity against Ba/F3-EGFR<sup>19del/T790M/C797S</sup> and Ba/F3-EGFR<sup>L858R/T790M/C797S</sup> cells with IC<sub>50</sub> values of 41 and 52 nM, respectively. Further, <b>21v</b> inhibited proliferation of the EGFR<sup>19del/T790M/C797S</sup> mutant PC-9-OR NSCLC cell line with an IC<sub>50</sub> value of 56 nM and displayed selectivity over parental Ba/F3 and A431 cells. Moreover, <b>21v</b> exhibited antitumor efficacy in a Ba/F3-EGFR<sup>19del/T790M/C797S</sup> xenograft model. This study provides a promising macrocyclic lead for anticancer drug discovery overcoming EGFR<sup>C797S</sup> mutation mediated resistance in NSCLC patients.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142609863","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}
{"title":"Proline Analogues in Drug Design: Current Trends and Future Prospects","authors":"Vladimir Kubyshkin, Pavel K. Mykhailiuk","doi":"10.1021/acs.jmedchem.4c01987","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01987","url":null,"abstract":"Proline analogues are versatile chemical building blocks that enable modular construction of small-molecule drugs and pharmaceutical peptides. Over the past 15 years, the FDA has approved over 15 drugs containing proline analogues in their structures, five in the last three years alone (daridorexant, trofinetide, nirmatrelvir, rezafungin, danicopan). This perspective offers an analysis of the most common types of proline analogues currently trending in drug design. We focus on examples of fluoroprolines, α-methylproline, bicyclic proline analogues, and aminoprolines, while also highlighting proline analogues that remain underrepresented. We supplement our analysis with physicochemical information regarding the specific molecular properties of these moieties. Additionally, we discuss several intriguing cases where nonproline residues were replaced with proline analogues as a strategy to eliminate unwanted hydrogen bond donor sites. In conclusion, we present some suggestions for the future exploration of this promising class of molecular entities in drug discovery.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142599575","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}
Xuejiao Shirley Guo, Sandeep Atla, Satyanarayana Nyalata, Yugendar R. Alugubelli, Peng-Hsun Chase Chen, Shiqing Xu, Wenshe Ray Liu
{"title":"Prioritization of Eleven-Nineteen-Leukemia Inhibitors as Orally Available Drug Candidates for Acute Myeloid Leukemia","authors":"Xuejiao Shirley Guo, Sandeep Atla, Satyanarayana Nyalata, Yugendar R. Alugubelli, Peng-Hsun Chase Chen, Shiqing Xu, Wenshe Ray Liu","doi":"10.1021/acs.jmedchem.4c01337","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01337","url":null,"abstract":"Acute myeloid leukemia (AML) is the second most prevalent and fatal form of leukemia. The growth of AML cells harboring oncogenic MLL rearrangements relies on the YEATS domain-containing protein ENL. Many small molecule inhibitors targeting ENL have been developed. To prioritize these inhibitors for in vivo studies, a NanoBRET system was introduced to evaluate their cellular permeability and potency. This screening identified inhibitor <b>13</b> as a promising candidate. This inhibitor has remarkable metabolic stability and potent antiproliferative effects on MLL-fusion leukemia cell lines. In AML-xenografted mice, inhibitor <b>13</b> significantly improved survival. Subsequent optimization efforts led to the development of <b>SR-C-107 (R)</b>, which exhibited strong activity against AML both at the cellular level (<i>CC</i><sub>50 (MOLM-13)</sub>: 1.25 ± 0.18 μM; <i>CC</i><sub>50 (MV4-11)</sub>: 0.81 ± 0.15 μM) and in vivo. These findings establish <b>SR-C-107 (R)</b> as a compelling candidate for AML treatment and lay the groundwork for the development of next-generation AML inhibitors.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142599573","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}
Giulia Alboreggia, Parima Udompholkul, Emma L. Atienza, Kendall Muzzarelli, Zahra Assar, Maurizio Pellecchia
{"title":"Covalent Targeting of Histidine Residues with Aryl Fluorosulfates: Application to Mcl-1 BH3 Mimetics","authors":"Giulia Alboreggia, Parima Udompholkul, Emma L. Atienza, Kendall Muzzarelli, Zahra Assar, Maurizio Pellecchia","doi":"10.1021/acs.jmedchem.4c01541","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01541","url":null,"abstract":"Covalent drugs provide pharmacodynamic and pharmacokinetic advantages over reversible agents. However, covalent strategies have been developed mostly to target cysteine (Cys) residues, which are rarely found in binding sites. Among other nucleophilic residues that could be in principle used for the design of covalent drugs, histidine (His) has not been given proper attention despite being in principle an attractive residue to pursue but underexplored. Aryl fluorosulfates, a mild electrophile that is very stable in biological media, have been recently identified as possible electrophiles to react with the side chains of Lys; however, limited studies are available on aryl fluorosulfates’ ability to target His residues. We demonstrate that proper incorporation of an aryl fluorosulfate juxtaposing the electrophile with a His residue can be used to afford rapid optimizations of His-covalent agents. As an application, we report on His-covalent BH3 mimetics targeting His224 of Mcl-1.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142601162","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}
Céline Delaitre, Michel Boisbrun, Samir Acherar, André Dias, Alexandra Kleinclauss, Mathilde Achard, Mélissa Colin, Trung Minh Nguyen, Nicolas Humbert, Khawla Chmeis, Karen L. Martinez, Nicolas Gilles, Philippe Robin, Sandra Lecat, François Dupuis
{"title":"Synthesis and Pharmacological Characterization of Fluorescent Ligands Targeting the Angiotensin II Receptors Derived from Agonists, β-Arrestin-Biased Agonists, and Antagonists","authors":"Céline Delaitre, Michel Boisbrun, Samir Acherar, André Dias, Alexandra Kleinclauss, Mathilde Achard, Mélissa Colin, Trung Minh Nguyen, Nicolas Humbert, Khawla Chmeis, Karen L. Martinez, Nicolas Gilles, Philippe Robin, Sandra Lecat, François Dupuis","doi":"10.1021/acs.jmedchem.4c01693","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c01693","url":null,"abstract":"Angiotensin II (AngII) regulates cerebral circulation and binds with a similar affinity to AT<sub>1</sub> and AT<sub>2</sub> receptors. Biased AT<sub>1</sub> agonists, such as TRV027, which are able to selectively activate β-arrestin while blocking the G<sub>q</sub> pathway, appear promising as new therapeutics. New pharmacological tools are needed to further explore the impact of biased AT<sub>1</sub> agonists on cells or tissues, such as the cerebral vessels. We designed and synthesized new fluorescent derivatives based on AngII, TRV027, or the AT<sub>1</sub> antagonist losartan. We conducted pharmacological characterization to determine their selectivity, potency, and ability to activate or not specific AT<sub>1</sub> transduction pathways in cells and cerebral arteries. We report the first highly AT<sub>1</sub>-selective fluorescent ligand, based on losartan, that retains its antagonist activity with high affinity. Fluorescent derivatives of TRV027 become AT<sub>2</sub>-selective and lose their AT<sub>1</sub> β-arrestin bias. These new ligands can be applied to trace AT<sub>1</sub> or AT<sub>2</sub> receptors in vitro and ex vivo.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142599040","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}
{"title":"Unnatural Amino Acids: Strategies, Designs, and Applications in Medicinal Chemistry and Drug Discovery","authors":"Krishna K. Sharma, Komal Sharma, Kamya Rao, Anku Sharma, Gajanan K. Rathod, Shams Aaghaz, Naina Sehra, Rajesh Parmar, Brett VanVeller, Rahul Jain","doi":"10.1021/acs.jmedchem.4c00110","DOIUrl":"https://doi.org/10.1021/acs.jmedchem.4c00110","url":null,"abstract":"Peptides can operate as therapeutic agents that sit within a privileged space between small molecules and larger biologics. Despite examples of their potential to regulate receptors and modulate disease pathways, the development of peptides with drug-like properties remains a challenge. In the quest to optimize physicochemical parameters and improve target selectivity, unnatural amino acids (UAAs) have emerged as critical tools in peptide- and peptidomimetic-based drugs. The utility of UAAs is illustrated by clinically approved drugs such as methyldopa, baclofen, and gabapentin in addition to small drug molecules, for example, bortezomib and sitagliptin. In this Perspective, we outline the strategy and deployment of UAAs in FDA-approved drugs and their targets. We further describe the modulation of the physicochemical properties in peptides using UAAs. Finally, we elucidate how these improved pharmacological parameters and the role played by UAAs impact the progress of analogs in preclinical stages with an emphasis on the role played by UAAs.","PeriodicalId":46,"journal":{"name":"Journal of Medicinal Chemistry","volume":null,"pages":null},"PeriodicalIF":7.3,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142599039","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}