Pub Date : 2024-08-17Epub Date: 2024-07-01DOI: 10.1080/17568919.2024.2366694
Dalal Nasser Binjawhar, Fawziah A Al-Salmi, Maha Ali Alghamdi, Ola A Abu Ali, Eman Fayad, Youstina William Rizzk, Nourhan M Ali, Ibrahim Mohey El-Deen, Elsayed H Eltamany
Aim: To synthesize new hybrid cinnamic acids (10a, 10b and 11) and ester derivatives (7, 8 and 9) and investigate their anti-breast cancer activities.Materials & methods: Compounds 7-11 were evaluated (in vitro) for their cytotoxic activities against the MCF-7 cell line. A flow cytometry examination was performed. Protein levels of nuclear factor erythroid 2-related factor 2 (Nrf2), topoisomerase II and caspase-9 were measured by qRT-PCR. Molecular docking studies were conducted.Results: Several components were discovered to be active, mainly component 11, which induced arrest in the cell cycle at phase S, greatly decreased the expression of Nrf2 and topoisomerase II; and upregulated the expression of caspase-9.Conclusion: The newly thiohydantoin-cinnamic acid hybrids can contribute to creating promising candidates for cancer drugs.
目的:合成新的混合肉桂酸(10a、10b 和 11)和酯衍生物(7、8 和 9),并研究它们的抗乳腺癌活性。材料与方法:在体外评估了化合物 7-11 对 MCF-7 细胞系的细胞毒性活性。进行了流式细胞术检测。通过 qRT-PCR 检测了核因子红细胞 2 相关因子 2(Nrf2)、拓扑异构酶 II 和 caspase-9 的蛋白水平。进行了分子对接研究。结果:发现了几种具有活性的成分,主要是成分 11,它诱导细胞周期停滞在 S 期,大大降低了 Nrf2 和拓扑异构酶 II 的表达,并上调了 caspase-9 的表达。结论新发现的硫代海因-肉桂酸杂交化合物有助于创造有前景的候选抗癌药物。
{"title":"<i>In vitro</i> anti-breast cancer study of hybrid cinnamic acid derivatives bearing 2-thiohydantoin moiety.","authors":"Dalal Nasser Binjawhar, Fawziah A Al-Salmi, Maha Ali Alghamdi, Ola A Abu Ali, Eman Fayad, Youstina William Rizzk, Nourhan M Ali, Ibrahim Mohey El-Deen, Elsayed H Eltamany","doi":"10.1080/17568919.2024.2366694","DOIUrl":"10.1080/17568919.2024.2366694","url":null,"abstract":"<p><p><b>Aim</b>: To synthesize new hybrid cinnamic acids (<b>10a</b>, <b>10b</b> and <b>11</b>) and ester derivatives (<b>7</b>, <b>8</b> and <b>9</b>) and investigate their anti-breast cancer activities.<b>Materials & methods:</b> Compounds <b>7-11</b> were evaluated (<i>in vitro</i>) for their cytotoxic activities against the MCF-7 cell line. A flow cytometry examination was performed. Protein levels of nuclear factor erythroid 2-related factor 2 (Nrf2), topoisomerase II and caspase-9 were measured by qRT-PCR. Molecular docking studies were conducted.<b>Results</b>: Several components were discovered to be active, mainly component <b>11</b>, which induced arrest in the cell cycle at phase S, greatly decreased the expression of Nrf2 and topoisomerase II; and upregulated the expression of caspase-9.<b>Conclusion:</b> The newly thiohydantoin-cinnamic acid hybrids can contribute to creating promising candidates for cancer drugs.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1665-1684"},"PeriodicalIF":3.2,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11370905/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141467402","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-17Epub Date: 2024-06-28DOI: 10.1080/17568919.2024.2366159
Shoaib Khan, Rafaqat Hussain, Tayyiaba Iqbal, Yousaf Khan, Urooj Jamal, Hany W Darwish, Muhammad Adnan
Aim: The present study describes benzothiazole derived thiazolidinone based thiadiazole derivatives (1-16) as anti-Alzheimer agents.Materials & methods: Synthesis of benzothiazole derived thiazolidinone based thiadiazole derivatives was achieved using the benzothiazole bearing 2-amine moiety. These synthesized compounds were confirmed via spectroscopic techniques (1H NMR, 13C NMR and HREI-MS). These compounds were biologically evaluated for their anti-Alzheimer potential. Binding interactions with proteins and drug likeness of the analogs were explored through molecular docking and ADMET analysis, respectively. In the novel series, compound-3 emerged as the most potent inhibitor when compared with other derivatives of the series.Conclusion: The present study provides potent anti-Alzheimer's agents that can be further optimized to discover novel anti-Alzheimer's drugs.
{"title":"Identification of novel benzothiazole-thiadiazole-based thiazolidinone derivative: <i>in vitro</i> and <i>in silico</i> approaches to develop promising anti-Alzheimer's agents.","authors":"Shoaib Khan, Rafaqat Hussain, Tayyiaba Iqbal, Yousaf Khan, Urooj Jamal, Hany W Darwish, Muhammad Adnan","doi":"10.1080/17568919.2024.2366159","DOIUrl":"10.1080/17568919.2024.2366159","url":null,"abstract":"<p><p><b>Aim:</b> The present study describes benzothiazole derived thiazolidinone based thiadiazole derivatives (<b>1-16</b>) as anti-Alzheimer agents.<b>Materials & methods:</b> Synthesis of benzothiazole derived thiazolidinone based thiadiazole derivatives was achieved using the benzothiazole bearing 2-amine moiety. These synthesized compounds were confirmed via spectroscopic techniques (<sup>1</sup>H NMR, <sup>13</sup>C NMR and HREI-MS). These compounds were biologically evaluated for their anti-Alzheimer potential. Binding interactions with proteins and drug likeness of the analogs were explored through molecular docking and ADMET analysis, respectively. In the novel series, compound-<b>3</b> emerged as the most potent inhibitor when compared with other derivatives of the series.<b>Conclusion:</b> The present study provides potent anti-Alzheimer's agents that can be further optimized to discover novel anti-Alzheimer's drugs.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1601-1613"},"PeriodicalIF":3.2,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11370920/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141467404","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-17Epub Date: 2024-07-16DOI: 10.1080/17568919.2024.2366158
Asmaa F Kassem, Mohamed A Omar, Ahmed Temirak, Riham A El-Shiekh, Aladdin M Srour
Aim: Design and synthesis of a series of 5-benzylidene(thio)barbiturates 3a-r.Methodology: Evaluation of the inhibitory activity of the new chemical entities on acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) using Donepezil as the standard reference.Results & Conclusion: Compound 3r emerged as the most potent AChE inhibitor (IC50 = 9.12 μM), while compound 3q exhibited the highest inhibitory activity against BChE (IC50 = 19.43 μM). Toxicological bioassays confirmed the absence of cytotoxicity for the most potent compounds at the tested doses. Molecular docking analysis demonstrated that the tested derivatives effectively bind to the active sites of both enzymes. Overall, this study sheds light on the potential of barbiturate-sulfonate conjugates as promising drug candidates.
{"title":"Barbiturate-sulfonate hybrids as potent cholinesterase inhibitors: design, synthesis and molecular modeling studies.","authors":"Asmaa F Kassem, Mohamed A Omar, Ahmed Temirak, Riham A El-Shiekh, Aladdin M Srour","doi":"10.1080/17568919.2024.2366158","DOIUrl":"10.1080/17568919.2024.2366158","url":null,"abstract":"<p><p><b>Aim:</b> Design and synthesis of a series of 5-benzylidene(thio)barbiturates <b>3a-r</b>.<b>Methodology:</b> Evaluation of the inhibitory activity of the new chemical entities on acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) using Donepezil as the standard reference.<b>Results & Conclusion:</b> Compound <b>3r</b> emerged as the most potent AChE inhibitor (IC<sub>50</sub> = 9.12 μM), while compound <b>3q</b> exhibited the highest inhibitory activity against BChE (IC<sub>50</sub> = 19.43 μM). Toxicological bioassays confirmed the absence of cytotoxicity for the most potent compounds at the tested doses. Molecular docking analysis demonstrated that the tested derivatives effectively bind to the active sites of both enzymes. Overall, this study sheds light on the potential of barbiturate-sulfonate conjugates as promising drug candidates.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1615-1631"},"PeriodicalIF":3.2,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11370902/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141619704","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-17Epub Date: 2024-08-05DOI: 10.1080/17568919.2024.2380245
Changyu Ren, Zhi Cao, Yang Liu, Rui Wang, Congcong Lin, Zishu Wang
Adiposity and obesity-related proteins (FTO), the earliest identified mRNA N6-methyladenosine (m6A) demethylases, are known to play crucial roles in several biological processes. Therefore, FTO is a promising target for anticancer treatment. Understanding the biological functions and regulatory mechanisms of FTO targets can serve as guidelines for drug development. Despite significant efforts to develop FTO inhibitors, no specific small-molecule inhibitors have entered clinical trials so far. In this manuscript, we review the relationship between FTO and various cancers, the small-molecule inhibitors developed against FTO targets from the perspective of medicinal chemistry and other fields, and describe their structural optimization process and structure-activity relationship, providing clues for their future development direction.
{"title":"Medicinal chemistry aspects of fat mass and obesity associated protein: structure, function and inhibitors.","authors":"Changyu Ren, Zhi Cao, Yang Liu, Rui Wang, Congcong Lin, Zishu Wang","doi":"10.1080/17568919.2024.2380245","DOIUrl":"10.1080/17568919.2024.2380245","url":null,"abstract":"<p><p>Adiposity and obesity-related proteins (FTO), the earliest identified mRNA <i>N</i><sup>6</sup>-methyladenosine (m<sup>6</sup>A) demethylases, are known to play crucial roles in several biological processes. Therefore, FTO is a promising target for anticancer treatment. Understanding the biological functions and regulatory mechanisms of FTO targets can serve as guidelines for drug development. Despite significant efforts to develop FTO inhibitors, no specific small-molecule inhibitors have entered clinical trials so far. In this manuscript, we review the relationship between FTO and various cancers, the small-molecule inhibitors developed against FTO targets from the perspective of medicinal chemistry and other fields, and describe their structural optimization process and structure-activity relationship, providing clues for their future development direction.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1705-1726"},"PeriodicalIF":3.2,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11370915/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141888966","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-02Epub Date: 2024-07-02DOI: 10.1080/17568919.2024.2359362
Muhammad Sajid, Hina Siddiqui, Humaira Zafar, Sammer Yousuf, Michael D Threadgill, Muhammad Iqbal Choudhary
Aim: We aim to develop new anti-leishmanial agents against Leishmania major and Leishmania tropica.Materials & methods: A total of 23 thiourea derivatives of (±)-aminoglutethimide were synthesized and evaluated for in vitro activity against promastigotes of L. major and L. tropica.Results & conclusion: The N-benzoyl analogue 7p was found potent (IC50 = 12.7 μM) against L. major and non toxic to normal cells. The docking studies, indicates that these inhibitors may target folate and glycolytic pathways of the parasite. The N-hexyl compound 7v was found strongly active against both species, and lacked cytotoxicity against normal cells, whereas compound 7r, with a 3,5-bis-(tri-fluoro-methyl)phenyl unit, was active against Leishmania, but was cytotoxic in nature. Compound 7v was thus identified as a hit for further studies.
{"title":"Thiourea-functionalized aminoglutethimide derivatives as anti-leishmanial agents.","authors":"Muhammad Sajid, Hina Siddiqui, Humaira Zafar, Sammer Yousuf, Michael D Threadgill, Muhammad Iqbal Choudhary","doi":"10.1080/17568919.2024.2359362","DOIUrl":"10.1080/17568919.2024.2359362","url":null,"abstract":"<p><p><b>Aim:</b> We aim to develop new anti-leishmanial agents against <i>Leishmania major</i> and <i>Leishmania tropica</i>.<b>Materials & methods:</b> A total of 23 thiourea derivatives of (±)-aminoglutethimide were synthesized and evaluated for <i>in vitro</i> activity against promastigotes of <i>L. major</i> and <i>L. tropica</i>.<b>Results & conclusion:</b> The <i>N</i>-benzoyl analogue <b>7p</b> was found potent (IC<sub>50</sub> = 12.7 μM) against <i>L. major</i> and non toxic to normal cells. The docking studies, indicates that these inhibitors may target folate and glycolytic pathways of the parasite. The <i>N</i>-hexyl compound <b>7v</b> was found strongly active against both species, and lacked cytotoxicity against normal cells, whereas compound <b>7r</b>, with a 3,5-bis-(tri-fluoro-methyl)phenyl unit, was active against <i>Leishmania</i>, but was cytotoxic in nature. Compound <b>7v</b> was thus identified as a hit for further studies.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1485-1497"},"PeriodicalIF":3.2,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11370960/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141491535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-02Epub Date: 2024-06-12DOI: 10.1080/17568919.2024.2359894
Diba Shareghi-Boroujeni, Aida Iraji, Mahintaj Dara, Mohammad Hashem Hashempur, Shahrokh Zare, Roshanak Hariri, Tahmineh Akbarzadeh, Mina Saeedi
Aim: A new series of 1,2,3-triazole-hydrazone derivatives were developed to evaluate their anti-Alzheimer's activity. Materials & methods: All compounds were screened toward cholinesterases via the modified Ellman's method. The toxicity assay on SH-SY5Y cells was performed using the MTT assay, and the expression levels of GSK-3α, GSK-3β, DYRK1 and CDK5 were assessed in the presence of compounds 6m and 6p.Results:6m and 6p; acting as mixed-type inhibitors, exhibited promising acetylcholinesterase and butyrylcholinesterase inhibitory activity, respectively. 6m demonstrated no toxicity under tested concentrations on the SH-SY5Y cells and positively impacted neurodegenerative pathways. Notably, 6m displayed a significant downregulation in mRNA levels of GSK-3α, GSK-3β and CDK5.Conclusion: The target compounds could be considered in developing anti-Alzheimer's disease agents.
{"title":"Synthesis of novel hybrids of 1,2,3-triazoles-hydrazone: targeting cholinesterases and Alzheimer's related genes.","authors":"Diba Shareghi-Boroujeni, Aida Iraji, Mahintaj Dara, Mohammad Hashem Hashempur, Shahrokh Zare, Roshanak Hariri, Tahmineh Akbarzadeh, Mina Saeedi","doi":"10.1080/17568919.2024.2359894","DOIUrl":"10.1080/17568919.2024.2359894","url":null,"abstract":"<p><p><b>Aim:</b> A new series of 1,2,3-triazole-hydrazone derivatives were developed to evaluate their anti-Alzheimer's activity. <b>Materials & methods:</b> All compounds were screened toward cholinesterases via the modified Ellman's method. The toxicity assay on SH-SY5Y cells was performed using the MTT assay, and the expression levels of <i>GSK-3α</i>, <i>GSK-3β</i>, <i>DYRK1</i> and <i>CDK5</i> were assessed in the presence of compounds <b>6m</b> and <b>6p</b>.<b>Results:6m</b> and <b>6p</b>; acting as mixed-type inhibitors, exhibited promising acetylcholinesterase and butyrylcholinesterase inhibitory activity, respectively. <b>6m</b> demonstrated no toxicity under tested concentrations on the SH-SY5Y cells and positively impacted neurodegenerative pathways. Notably, <b>6m</b> displayed a significant downregulation in mRNA levels of <i>GSK-3α</i>, <i>GSK-3β</i> and <i>CDK5</i>.<b>Conclusion:</b> The target compounds could be considered in developing anti-Alzheimer's disease agents.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1519-1535"},"PeriodicalIF":3.2,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11370907/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141305754","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-02Epub Date: 2024-07-01DOI: 10.1080/17568919.2024.2366690
Lívia Cristina de Souza Viol, Natália Aparecida Liberto Silva, Cristiane Isaac Cerceau, Marcus Vinícius de Andrade Barros, Raoni Pais Siqueira, Victor Hugo Sousa Gonçalves, Gustavo Costa Bressan, Sergio Antonio Fernandes, Elson Santiago Alvarenga, Róbson Ricardo Teixeira
Aim: This study aimed to enhance the aqueous dissolution of SRPK inhibitor N-(2-(piperidin-1-yl)-5-(trifluoromethyl)phenyl)isonicotinamide (SRPIN340).Materials & Methods: A complex with p-sulfonic calix[6]arene (Host) and SRPIN340 (Guest) was prepared, studied via 1H nuclear magnetic resonance (NMR) and theoretical calculations and biologically evaluated on cancer cell lines.Results & conclusion: The 1:1 host (H)/guest (G) complex significantly enhanced the aqueous dissolution of SRPIN340, achieving 64.8% water solubility as determined by 1H NMR quantification analysis. The H/G complex reduced cell viability by 75% for HL60, ∼50% for Nalm6 and Jurkat, and ∼30% for B16F10 cells. It exhibited greater cytotoxicity than free SRPIN340 against Jurkat and B16F10 cells. Theoretical studies indicated hydrogen bond stabilization of the complex, suggesting broader applicability of SRPIN340 across diverse biological systems.
{"title":"NMR analysis, cytotoxic activity and theoretical study of a complex between SRPIN340 and <i>p</i>-sulfonic acid calix[6]arene.","authors":"Lívia Cristina de Souza Viol, Natália Aparecida Liberto Silva, Cristiane Isaac Cerceau, Marcus Vinícius de Andrade Barros, Raoni Pais Siqueira, Victor Hugo Sousa Gonçalves, Gustavo Costa Bressan, Sergio Antonio Fernandes, Elson Santiago Alvarenga, Róbson Ricardo Teixeira","doi":"10.1080/17568919.2024.2366690","DOIUrl":"10.1080/17568919.2024.2366690","url":null,"abstract":"<p><p><b>Aim:</b> This study aimed to enhance the aqueous dissolution of SRPK inhibitor <i>N</i>-(2-(piperidin-1-yl)-5-(trifluoromethyl)phenyl)isonicotinamide (SRPIN340).<b>Materials & Methods:</b> A complex with <i>p</i>-sulfonic calix[6]arene (Host) and SRPIN340 (Guest) was prepared, studied via <sup>1</sup>H nuclear magnetic resonance (NMR) and theoretical calculations and biologically evaluated on cancer cell lines.<b>Results & conclusion:</b> The 1:1 host (H)/guest (G) complex significantly enhanced the aqueous dissolution of SRPIN340, achieving 64.8% water solubility as determined by <sup>1</sup>H NMR quantification analysis. The H/G complex reduced cell viability by 75% for HL60, ∼50% for Nalm6 and Jurkat, and ∼30% for B16F10 cells. It exhibited greater cytotoxicity than free SRPIN340 against Jurkat and B16F10 cells. Theoretical studies indicated hydrogen bond stabilization of the complex, suggesting broader applicability of SRPIN340 across diverse biological systems.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1537-1550"},"PeriodicalIF":3.2,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11370924/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141467405","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-02Epub Date: 2024-07-01DOI: 10.1080/17568919.2024.2363668
Nur Farisya Shamsudin, Sze-Wei Leong, Andreas Koeberle, Utid Suriya, Thanyada Rungrotmongkol, Suet Lin Chia, Muhammad Taher, Muhammad Salahuddin Haris, Hussah Abdullah Alshwyeh, Areej A Alosaimi, Ahmed Mediani, Muna Abdulsalam Ilowefah, Deri Islami, Siti Munirah Mohd Faudzi, Mohd Fadhlizil Fasihi Mohd Aluwi, Lam Kok Wai, Kamal Rullah
Aim: Chromones are promising for anticancer drug development.Methods & results: 12 chromone-based compounds were synthesized and tested against cancer cell lines. Compound 8 showed the highest cytotoxicity (LC50 3.2 μM) against colorectal cancer cells, surpassing 5-fluorouracil (LC50 4.2 μM). It suppressed colony formation, induced cell cycle arrest and triggered apoptotic cell death, confirmed by staining and apoptosis markers. Cell death was accompanied by enhanced reactive oxygen species formation and modulation of the autophagic machinery (autophagy marker light chain 3B (LC3B); adenosine monophosphate-activated protein kinase (AMPK); protein kinase B (PKB); UNC-51-like kinase (ULK)-1; and ULK2). Molecular docking and dynamic simulations revealed that compound 8 directly binds to ULK1.Conclusion: Compound 8 is a promising lead for autophagy-modulating anti-colon cancer drugs.
{"title":"A novel chromone-based as a potential inhibitor of ULK1 that modulates autophagy and induces apoptosis in colon cancer.","authors":"Nur Farisya Shamsudin, Sze-Wei Leong, Andreas Koeberle, Utid Suriya, Thanyada Rungrotmongkol, Suet Lin Chia, Muhammad Taher, Muhammad Salahuddin Haris, Hussah Abdullah Alshwyeh, Areej A Alosaimi, Ahmed Mediani, Muna Abdulsalam Ilowefah, Deri Islami, Siti Munirah Mohd Faudzi, Mohd Fadhlizil Fasihi Mohd Aluwi, Lam Kok Wai, Kamal Rullah","doi":"10.1080/17568919.2024.2363668","DOIUrl":"10.1080/17568919.2024.2363668","url":null,"abstract":"<p><p><b>Aim:</b> Chromones are promising for anticancer drug development.<b>Methods & results:</b> 12 chromone-based compounds were synthesized and tested against cancer cell lines. Compound <b>8</b> showed the highest cytotoxicity (LC<sub>50</sub> 3.2 μM) against colorectal cancer cells, surpassing 5-fluorouracil (LC<sub>50</sub> 4.2 μM). It suppressed colony formation, induced cell cycle arrest and triggered apoptotic cell death, confirmed by staining and apoptosis markers. Cell death was accompanied by enhanced reactive oxygen species formation and modulation of the autophagic machinery (autophagy marker light chain 3B (LC3B); adenosine monophosphate-activated protein kinase (AMPK); protein kinase B (PKB); UNC-51-like kinase (ULK)-1; and ULK2). Molecular docking and dynamic simulations revealed that compound <b>8</b> directly binds to ULK1.<b>Conclusion:</b> Compound <b>8</b> is a promising lead for autophagy-modulating anti-colon cancer drugs.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1499-1517"},"PeriodicalIF":3.2,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11370956/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141467403","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-02Epub Date: 2024-07-01DOI: 10.1080/17568919.2024.2366146
Jinglin Tang, Han Liu, Jinze Li, Yibo Zhang, Suyang Yao, Kan Yang, Zhihao You, Xiaoqiang Qiao, Yali Song
PD-L1 is overexpressed on the surface of tumor cells and binds to PD-1, resulting in tumor immune escape. Therapeutic strategies to target the PD-1/PD-L1 pathway involve blocking the binding. Immune checkpoint inhibitors have limited efficacy against tumors because PD-L1 is also present in the cytoplasm. PD-L1 of post-translational modifications (PTMs) have uncovered numerous mechanisms contributing to carcinogenesis and have identified potential therapeutic targets. Therefore, small molecule inhibitors can block crucial carcinogenic signaling pathways, making them a potential therapeutic option. To better develop small molecule inhibitors, we have summarized the PTMs of PD-L1. This review discusses the regulatory mechanisms of small molecule inhibitors in carcinogenesis and explore their potential applications, proposing a novel approach for tumor immunotherapy based on PD-L1 PTM.
{"title":"Regulation of post-translational modification of PD-L1 and associated opportunities for novel small-molecule therapeutics.","authors":"Jinglin Tang, Han Liu, Jinze Li, Yibo Zhang, Suyang Yao, Kan Yang, Zhihao You, Xiaoqiang Qiao, Yali Song","doi":"10.1080/17568919.2024.2366146","DOIUrl":"10.1080/17568919.2024.2366146","url":null,"abstract":"<p><p>PD-L1 is overexpressed on the surface of tumor cells and binds to PD-1, resulting in tumor immune escape. Therapeutic strategies to target the PD-1/PD-L1 pathway involve blocking the binding. Immune checkpoint inhibitors have limited efficacy against tumors because PD-L1 is also present in the cytoplasm. PD-L1 of post-translational modifications (PTMs) have uncovered numerous mechanisms contributing to carcinogenesis and have identified potential therapeutic targets. Therefore, small molecule inhibitors can block crucial carcinogenic signaling pathways, making them a potential therapeutic option. To better develop small molecule inhibitors, we have summarized the PTMs of PD-L1. This review discusses the regulatory mechanisms of small molecule inhibitors in carcinogenesis and explore their potential applications, proposing a novel approach for tumor immunotherapy based on PD-L1 PTM.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1583-1599"},"PeriodicalIF":3.2,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11370925/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141467406","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-08-02Epub Date: 2024-06-20DOI: 10.1080/17568919.2024.2363672
Ahmed Bm Ibrahim, Ereny S Williem, S Abd Elkhalik, Alexander Villinger, S M Abbas
Aim: Zinc salicylaldimines may act as multidrug agents.Results: Three zinc salicylaldimines C1-C3 and respective ligands HL1-HL3 were examined for antimicrobial/anticancer drug action and C3 was structurally analyzed (tetrahedral, triclinic). Against two fungi, C1 inhibited Candida albicans with 12 mm (21 mm for amphotericin B). Among four bacteria, two ligands inhibited Staphylococcus aureus and Escherichia coli (9-10 mm), but the complexes inhibited all bacteria with 10-14 mm (21-26 mm for ampicillin). The half-maximal inhibitory concentrations for the ligands, complexes and doxorubicin were 195.5-310.7, 22.18-70.05 and 9.66 μM against cancerous MCF-7 cells and 186.4-199.9, 14.95-18.87 and 36.42 μM against normal BHK cells.Conclusion: The complexation produced pronounced enhancement in the ligand antimicrobial/anticancer activities, despite these activities are moderate comparing with standards.
{"title":"Structural investigations and antibacterial, antifungal and anticancer studies on zinc salicylaldimine complexes.","authors":"Ahmed Bm Ibrahim, Ereny S Williem, S Abd Elkhalik, Alexander Villinger, S M Abbas","doi":"10.1080/17568919.2024.2363672","DOIUrl":"10.1080/17568919.2024.2363672","url":null,"abstract":"<p><p><b>Aim:</b> Zinc salicylaldimines may act as multidrug agents.<b>Results:</b> Three zinc salicylaldimines <b>C1-C3</b> and respective ligands <b>HL<sup>1</sup></b>-<b>HL<sup>3</sup></b> were examined for antimicrobial/anticancer drug action and <b>C3</b> was structurally analyzed (tetrahedral, triclinic). Against two fungi, <b>C1</b> inhibited <i>Candida albicans</i> with 12 mm (21 mm for amphotericin B). Among four bacteria, two ligands inhibited <i>Staphylococcus aureus</i> and <i>Escherichia coli</i> (9-10 mm), but the complexes inhibited all bacteria with 10-14 mm (21-26 mm for ampicillin). The half-maximal inhibitory concentrations for the ligands, complexes and doxorubicin were 195.5-310.7, 22.18-70.05 and 9.66 μM against cancerous MCF-7 cells and 186.4-199.9, 14.95-18.87 and 36.42 μM against normal BHK cells.<b>Conclusion:</b> The complexation produced pronounced enhancement in the ligand antimicrobial/anticancer activities, despite these activities are moderate comparing with standards.</p>","PeriodicalId":12475,"journal":{"name":"Future medicinal chemistry","volume":" ","pages":"1551-1560"},"PeriodicalIF":3.2,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11370977/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141426679","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}