Pub Date : 2024-10-15eCollection Date: 2024-11-14DOI: 10.1021/acsmedchemlett.4c00490
Robert B Kargbo
Recent advancements in the therapeutic use of psychedelic compounds, particularly 5-MeO-DMT and DMT, have demonstrated significant potential in treating various mental health disorders. This Patent Highlight evaluates findings from multiple recent patents that explore novel applications of these compounds, including their use in severe psychiatric conditions such as depression, anxiety, and trauma-related disorders. The patents reveal innovative strategies, such as combining 5-MeO-DMT with mood preparation agents to enhance safety and efficacy, developing DMT prodrugs for improved pharmacokinetics, and targeting specific populations like breastfeeding mothers with tailored psychedelic therapies. These advancements offer new hope for patients with treatment-resistant mental disorders, highlighting the evolving role of psychedelics in mental health care.
{"title":"Innovative Psychedelic Therapies: Harnessing 5-MeO-DMT and DMT for Mental Health Treatment.","authors":"Robert B Kargbo","doi":"10.1021/acsmedchemlett.4c00490","DOIUrl":"10.1021/acsmedchemlett.4c00490","url":null,"abstract":"<p><p>Recent advancements in the therapeutic use of psychedelic compounds, particularly 5-MeO-DMT and DMT, have demonstrated significant potential in treating various mental health disorders. This Patent Highlight evaluates findings from multiple recent patents that explore novel applications of these compounds, including their use in severe psychiatric conditions such as depression, anxiety, and trauma-related disorders. The patents reveal innovative strategies, such as combining 5-MeO-DMT with mood preparation agents to enhance safety and efficacy, developing DMT prodrugs for improved pharmacokinetics, and targeting specific populations like breastfeeding mothers with tailored psychedelic therapies. These advancements offer new hope for patients with treatment-resistant mental disorders, highlighting the evolving role of psychedelics in mental health care.</p>","PeriodicalId":20,"journal":{"name":"ACS Medicinal Chemistry Letters","volume":"15 11","pages":"1812-1814"},"PeriodicalIF":3.5,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11571087/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142674544","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-15eCollection Date: 2024-11-14DOI: 10.1021/acsmedchemlett.4c00488
Robert B Kargbo
Psilocybin, a naturally occurring psychedelic compound, has recently emerged as a promising therapeutic agent for mental health. This Patent Highlight explores the innovative approaches to harnessing psilocybin's potential across various contexts. These include clinical trials targeting severe psychiatric conditions, the integration of low-dose psilocybin into dietary products to promote general mental well-being, and the personalization of psilocybin dosing for optimal treatment of depression and anxiety. These advancements demonstrate psilocybin's versatility and potential to reshape conventional mental health treatment paradigms, mainly through personalized medicine and accessible wellness applications.
{"title":"Expanding the Therapeutic Horizons of Psilocybin in Mental Health.","authors":"Robert B Kargbo","doi":"10.1021/acsmedchemlett.4c00488","DOIUrl":"10.1021/acsmedchemlett.4c00488","url":null,"abstract":"<p><p>Psilocybin, a naturally occurring psychedelic compound, has recently emerged as a promising therapeutic agent for mental health. This Patent Highlight explores the innovative approaches to harnessing psilocybin's potential across various contexts. These include clinical trials targeting severe psychiatric conditions, the integration of low-dose psilocybin into dietary products to promote general mental well-being, and the personalization of psilocybin dosing for optimal treatment of depression and anxiety. These advancements demonstrate psilocybin's versatility and potential to reshape conventional mental health treatment paradigms, mainly through personalized medicine and accessible wellness applications.</p>","PeriodicalId":20,"journal":{"name":"ACS Medicinal Chemistry Letters","volume":"15 11","pages":"1821-1823"},"PeriodicalIF":3.5,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11571021/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142674528","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-15eCollection Date: 2024-11-14DOI: 10.1021/acsmedchemlett.4c00350
Natalie Fuchs, Laura Calvo-Barreiro, Valerij Talagayev, Szymon Pach, Gerhard Wolber, Moustafa T Gabr
Herein, we performed a virtual screening study to discover new scaffolds for small molecule-based ligands of the immune checkpoint lymphocyte-activation gene 3 (LAG-3). Molecular dynamics (MD) simulations using the LAG-3 structure revealed two putative binding sites for small molecules: the antibody interface and the lipophilic canyon. A 3D pharmacophore screening resulted in the identification of potential ligands for these binding sites and afforded a library of 25 compounds. We then evaluated the screening hits for LAG-3 binding via microscale thermophoresis (MST) and surface plasmon resonance (SPR). Our biophysical screening identified two binders with KD values in the low micromolar range, compounds 3 (antibody interface) and 25 (lipophilic canyon). Furthermore, we investigated the ability of LAG-3 hits to engage LAG-3 on a cellular level using a cellular thermal shift assay (CETSA). In summary, compound 3 shows potential as a lead but is not yet a development candidate.
在此,我们进行了一项虚拟筛选研究,以发现基于小分子的免疫检查点淋巴细胞活化基因 3(LAG-3)配体的新支架。利用 LAG-3 结构进行的分子动力学(MD)模拟揭示了小分子的两个潜在结合位点:抗体界面和亲油峡谷。通过三维药理筛选,我们确定了这些结合位点的潜在配体,并建立了一个由 25 种化合物组成的化合物库。然后,我们通过微尺度热泳(MST)和表面等离子体共振(SPR)评估了筛选出的 LAG-3 结合情况。我们的生物物理筛选确定了两种 K D 值在低微摩尔范围内的结合剂,即化合物 3(抗体界面)和 25(亲油峡谷)。此外,我们还利用细胞热转移试验(CETSA)研究了 LAG-3 命中化合物在细胞水平上与 LAG-3 结合的能力。总之,化合物 3 显示出作为先导物的潜力,但尚未成为候选开发物。
{"title":"From Virtual Screens to Cellular Target Engagement: New Small Molecule Ligands for the Immune Checkpoint LAG-3.","authors":"Natalie Fuchs, Laura Calvo-Barreiro, Valerij Talagayev, Szymon Pach, Gerhard Wolber, Moustafa T Gabr","doi":"10.1021/acsmedchemlett.4c00350","DOIUrl":"10.1021/acsmedchemlett.4c00350","url":null,"abstract":"<p><p>Herein, we performed a virtual screening study to discover new scaffolds for small molecule-based ligands of the immune checkpoint lymphocyte-activation gene 3 (LAG-3). Molecular dynamics (MD) simulations using the LAG-3 structure revealed two putative binding sites for small molecules: the antibody interface and the lipophilic canyon. A 3D pharmacophore screening resulted in the identification of potential ligands for these binding sites and afforded a library of 25 compounds. We then evaluated the screening hits for LAG-3 binding via microscale thermophoresis (MST) and surface plasmon resonance (SPR). Our biophysical screening identified two binders with <i>K</i> <sub>D</sub> values in the low micromolar range, compounds <b>3</b> (antibody interface) and <b>25</b> (lipophilic canyon). Furthermore, we investigated the ability of LAG-3 hits to engage LAG-3 on a cellular level using a cellular thermal shift assay (CETSA). In summary, compound <b>3</b> shows potential as a lead but is not yet a development candidate.</p>","PeriodicalId":20,"journal":{"name":"ACS Medicinal Chemistry Letters","volume":"15 11","pages":"1884-1890"},"PeriodicalIF":3.5,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11571005/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142674536","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-15DOI: 10.1021/acsmedchemlett.4c0048910.1021/acsmedchemlett.4c00489
Robert B. Kargbo*,
In the rapidly evolving field of oncology, the need for innovative therapeutic strategies is more pressing than ever. Many cancers, especially those driven by gene mutations like KRAS, remain challenging to treat. Traditional approaches, which often rely on small-molecule inhibitors, face significant limitations, particularly in addressing “undruggable” targets or delivering large therapeutic molecules across cellular membranes. As highlighted by four essential patents, the recent advancements in Proteolysis Targeting Chimeras (PROTACs) and intracellular delivery systems offer novel and promising strategies to overcome these challenges.
{"title":"A New Frontier in Targeted Therapies: Harnessing PROTACs and Advanced Delivery Systems","authors":"Robert B. Kargbo*, ","doi":"10.1021/acsmedchemlett.4c0048910.1021/acsmedchemlett.4c00489","DOIUrl":"https://doi.org/10.1021/acsmedchemlett.4c00489https://doi.org/10.1021/acsmedchemlett.4c00489","url":null,"abstract":"<p >In the rapidly evolving field of oncology, the need for innovative therapeutic strategies is more pressing than ever. Many cancers, especially those driven by gene mutations like KRAS, remain challenging to treat. Traditional approaches, which often rely on small-molecule inhibitors, face significant limitations, particularly in addressing “undruggable” targets or delivering large therapeutic molecules across cellular membranes. As highlighted by four essential patents, the recent advancements in Proteolysis Targeting Chimeras (PROTACs) and intracellular delivery systems offer novel and promising strategies to overcome these challenges.</p>","PeriodicalId":20,"journal":{"name":"ACS Medicinal Chemistry Letters","volume":"15 11","pages":"1818–1820 1818–1820"},"PeriodicalIF":3.5,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142641120","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-15DOI: 10.1021/acsmedchemlett.4c0048710.1021/acsmedchemlett.4c00487
Robert B. Kargbo*,
This Patent Highlight explores recent cancer treatment and monitoring advancements, focusing on selective KRAS inhibitors targeting mutations like G12C and G12D alongside germline epitope burden analysis. These innovations offer enhanced therapeutic precision and personalized monitoring, improving the prediction of cancer relapse and tailoring treatment strategies to individual patient profiles, significantly advancing the field of precision oncology.
{"title":"Advances in Cancer Treatment and Monitoring: Insights from KRAS Inhibitors and Germline Epitope Burden Monitoring","authors":"Robert B. Kargbo*, ","doi":"10.1021/acsmedchemlett.4c0048710.1021/acsmedchemlett.4c00487","DOIUrl":"https://doi.org/10.1021/acsmedchemlett.4c00487https://doi.org/10.1021/acsmedchemlett.4c00487","url":null,"abstract":"<p >This Patent Highlight explores recent cancer treatment and monitoring advancements, focusing on selective KRAS inhibitors targeting mutations like G12C and G12D alongside germline epitope burden analysis. These innovations offer enhanced therapeutic precision and personalized monitoring, improving the prediction of cancer relapse and tailoring treatment strategies to individual patient profiles, significantly advancing the field of precision oncology.</p>","PeriodicalId":20,"journal":{"name":"ACS Medicinal Chemistry Letters","volume":"15 11","pages":"1815–1817 1815–1817"},"PeriodicalIF":3.5,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142641118","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-15DOI: 10.1021/acsmedchemlett.4c0028910.1021/acsmedchemlett.4c00289
Joydip Mondal, Tiasha Dasgupta, Rakesh R. Panicker, Venkatraman Manickam, Arup Sinha and Akella Sivaramakrishna*,
Inhibition of vascular endothelial growth factor receptor 2 (VEGFR-2) facilitates potent antiangiogenic and anticancer responses. In this regard, the development of effective pharmacophores, i.e., quinoline-based triazole derivatives 6a–j, by a one-pot telescopic approach is our focus. Among all of them, 6f, possessing amide and cyanide substituents, displayed the highest binding ability with VEGFR-2, having high affinity of −8.9 kcal/mol. Further, 6f and 6g (containing amide and bromo groups) exhibited a wide spectrum of anticancer activities due to the presence of active oxidative stress inducers, with cytotoxicity values of 10 ± 0.2 and 12 ± 0.6 μM, respectively. Apoptosis analysis demonstrated the involvement of 6f and 6g in mitochondrial damage and the loss of mitochondrial membrane potential (ΔΨm). Intercellular localization of 6f/6g in MCF-7 revealed the presence of 6g in the cytoplasm along with an increase in ROS production and a reduction in MMP, proving the ability of 6g to target mitochondria.
{"title":"Promoting Apoptosis in MCF-7 Cells via ROS Generation by Quinolino-triazoles Derived from One-Pot Telescopic Synthesis","authors":"Joydip Mondal, Tiasha Dasgupta, Rakesh R. Panicker, Venkatraman Manickam, Arup Sinha and Akella Sivaramakrishna*, ","doi":"10.1021/acsmedchemlett.4c0028910.1021/acsmedchemlett.4c00289","DOIUrl":"https://doi.org/10.1021/acsmedchemlett.4c00289https://doi.org/10.1021/acsmedchemlett.4c00289","url":null,"abstract":"<p >Inhibition of vascular endothelial growth factor receptor 2 (VEGFR-2) facilitates potent antiangiogenic and anticancer responses. In this regard, the development of effective pharmacophores, i.e., quinoline-based triazole derivatives <b>6a</b>–<b>j</b>, by a one-pot telescopic approach is our focus. Among all of them, <b>6f</b>, possessing amide and cyanide substituents, displayed the highest binding ability with VEGFR-2, having high affinity of −8.9 kcal/mol. Further, <b>6f</b> and <b>6g</b> (containing amide and bromo groups) exhibited a wide spectrum of anticancer activities due to the presence of active oxidative stress inducers, with cytotoxicity values of 10 ± 0.2 and 12 ± 0.6 μM, respectively. Apoptosis analysis demonstrated the involvement of <b>6f</b> and <b>6g</b> in mitochondrial damage and the loss of mitochondrial membrane potential (ΔΨ<sub>m</sub>). Intercellular localization of <b>6f/6g</b> in MCF-7 revealed the presence of <b>6g</b> in the cytoplasm along with an increase in ROS production and a reduction in MMP, proving the ability of <b>6g</b> to target mitochondria.</p>","PeriodicalId":20,"journal":{"name":"ACS Medicinal Chemistry Letters","volume":"15 11","pages":"1866–1874 1866–1874"},"PeriodicalIF":3.5,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142641112","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-15DOI: 10.1021/acsmedchemlett.4c0048810.1021/acsmedchemlett.4c00488
Robert B. Kargbo*,
Psilocybin, a naturally occurring psychedelic compound, has recently emerged as a promising therapeutic agent for mental health. This Patent Highlight explores the innovative approaches to harnessing psilocybin’s potential across various contexts. These include clinical trials targeting severe psychiatric conditions, the integration of low-dose psilocybin into dietary products to promote general mental well-being, and the personalization of psilocybin dosing for optimal treatment of depression and anxiety. These advancements demonstrate psilocybin’s versatility and potential to reshape conventional mental health treatment paradigms, mainly through personalized medicine and accessible wellness applications.
{"title":"Expanding the Therapeutic Horizons of Psilocybin in Mental Health","authors":"Robert B. Kargbo*, ","doi":"10.1021/acsmedchemlett.4c0048810.1021/acsmedchemlett.4c00488","DOIUrl":"https://doi.org/10.1021/acsmedchemlett.4c00488https://doi.org/10.1021/acsmedchemlett.4c00488","url":null,"abstract":"<p >Psilocybin, a naturally occurring psychedelic compound, has recently emerged as a promising therapeutic agent for mental health. This Patent Highlight explores the innovative approaches to harnessing psilocybin’s potential across various contexts. These include clinical trials targeting severe psychiatric conditions, the integration of low-dose psilocybin into dietary products to promote general mental well-being, and the personalization of psilocybin dosing for optimal treatment of depression and anxiety. These advancements demonstrate psilocybin’s versatility and potential to reshape conventional mental health treatment paradigms, mainly through personalized medicine and accessible wellness applications.</p>","PeriodicalId":20,"journal":{"name":"ACS Medicinal Chemistry Letters","volume":"15 11","pages":"1821–1823 1821–1823"},"PeriodicalIF":3.5,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142641119","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-14DOI: 10.1021/acsmedchemlett.4c0027410.1021/acsmedchemlett.4c00274
Hui Sun, Cong Wang, Xiaona Li, Zirui Lü, Kebin Li, Hengjie Hu, Ping Xu, Yu Xiao* and Yan Niu*,
Targeted protein degradation has been emerging as a promising strategy for drug design and a useful tool for the research of intracellular protein function by specifically downregulating the protein level via promoted degradation. Aside from proteolysis targeting chimeras (PROTAC) that utilize a specific E3 ligase ligand as a tag to recruit polyubiquitin onto the targeted protein and subsequently induce degradation, Boc3Arg was also reported an efficient tag to induce degradation through directly localizing the protein to the 20S proteasome. Based on the similarity of Boc2Lys and Boc3Arg, we identified that Boc2Lys also efficiently induced targeted protein degradation, taking glutathione S-transferase as an example. We found that Boc2Lys-linked ethacrynic acid was able to dose-dependently downregulate the target protein in a mechanism distinct to Boc3Arg.
{"title":"Identification of Boc2Lys-Linked Ethacrynic Acid and Its Analogues As Efficient Glutathione S-Transferase Degraders","authors":"Hui Sun, Cong Wang, Xiaona Li, Zirui Lü, Kebin Li, Hengjie Hu, Ping Xu, Yu Xiao* and Yan Niu*, ","doi":"10.1021/acsmedchemlett.4c0027410.1021/acsmedchemlett.4c00274","DOIUrl":"https://doi.org/10.1021/acsmedchemlett.4c00274https://doi.org/10.1021/acsmedchemlett.4c00274","url":null,"abstract":"<p >Targeted protein degradation has been emerging as a promising strategy for drug design and a useful tool for the research of intracellular protein function by specifically downregulating the protein level via promoted degradation. Aside from proteolysis targeting chimeras (PROTAC) that utilize a specific E3 ligase ligand as a tag to recruit polyubiquitin onto the targeted protein and subsequently induce degradation, Boc<sub>3</sub>Arg was also reported an efficient tag to induce degradation through directly localizing the protein to the 20S proteasome. Based on the similarity of Boc<sub>2</sub>Lys and Boc<sub>3</sub>Arg, we identified that Boc<sub>2</sub>Lys also efficiently induced targeted protein degradation, taking glutathione S-transferase as an example. We found that Boc<sub>2</sub>Lys-linked ethacrynic acid was able to dose-dependently downregulate the target protein in a mechanism distinct to Boc<sub>3</sub>Arg.</p>","PeriodicalId":20,"journal":{"name":"ACS Medicinal Chemistry Letters","volume":"15 11","pages":"1852–1859 1852–1859"},"PeriodicalIF":3.5,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142641039","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-10-14DOI: 10.1021/acsmedchemlett.4c0048510.1021/acsmedchemlett.4c00485
Ram W. Sabnis*,
Provided herein are novel pyrazolopyridine and triazolopyridine derivatives as DGAT2 inhibitors, pharmaceutical compositions, use of such compounds in treating multiple diseases, and processes for preparing such compounds.
{"title":"Pyrazolopyridine and Triazolopyridine Derivatives as DGAT2 Inhibitors for Treating Multiple Diseases","authors":"Ram W. Sabnis*, ","doi":"10.1021/acsmedchemlett.4c0048510.1021/acsmedchemlett.4c00485","DOIUrl":"https://doi.org/10.1021/acsmedchemlett.4c00485https://doi.org/10.1021/acsmedchemlett.4c00485","url":null,"abstract":"<p >Provided herein are novel pyrazolopyridine and triazolopyridine derivatives as DGAT2 inhibitors, pharmaceutical compositions, use of such compounds in treating multiple diseases, and processes for preparing such compounds.</p>","PeriodicalId":20,"journal":{"name":"ACS Medicinal Chemistry Letters","volume":"15 11","pages":"1806–1807 1806–1807"},"PeriodicalIF":3.5,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142641072","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Targeted protein degradation has been emerging as a promising strategy for drug design and a useful tool for the research of intracellular protein function by specifically downregulating the protein level via promoted degradation. Aside from proteolysis targeting chimeras (PROTAC) that utilize a specific E3 ligase ligand as a tag to recruit polyubiquitin onto the targeted protein and subsequently induce degradation, Boc3Arg was also reported an efficient tag to induce degradation through directly localizing the protein to the 20S proteasome. Based on the similarity of Boc2Lys and Boc3Arg, we identified that Boc2Lys also efficiently induced targeted protein degradation, taking glutathione S-transferase as an example. We found that Boc2Lys-linked ethacrynic acid was able to dose-dependently downregulate the target protein in a mechanism distinct to Boc3Arg.
{"title":"Identification of Boc<sub>2</sub>Lys-Linked Ethacrynic Acid and Its Analogues As Efficient Glutathione S-Transferase Degraders.","authors":"Hui Sun, Cong Wang, Xiaona Li, Zirui Lü, Kebin Li, Hengjie Hu, Ping Xu, Yu Xiao, Yan Niu","doi":"10.1021/acsmedchemlett.4c00274","DOIUrl":"10.1021/acsmedchemlett.4c00274","url":null,"abstract":"<p><p>Targeted protein degradation has been emerging as a promising strategy for drug design and a useful tool for the research of intracellular protein function by specifically downregulating the protein level via promoted degradation. Aside from proteolysis targeting chimeras (PROTAC) that utilize a specific E3 ligase ligand as a tag to recruit polyubiquitin onto the targeted protein and subsequently induce degradation, Boc<sub>3</sub>Arg was also reported an efficient tag to induce degradation through directly localizing the protein to the 20S proteasome. Based on the similarity of Boc<sub>2</sub>Lys and Boc<sub>3</sub>Arg, we identified that Boc<sub>2</sub>Lys also efficiently induced targeted protein degradation, taking glutathione S-transferase as an example. We found that Boc<sub>2</sub>Lys-linked ethacrynic acid was able to dose-dependently downregulate the target protein in a mechanism distinct to Boc<sub>3</sub>Arg.</p>","PeriodicalId":20,"journal":{"name":"ACS Medicinal Chemistry Letters","volume":"15 11","pages":"1852-1859"},"PeriodicalIF":3.5,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11571028/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142674542","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}