{"title":"Phoenixin-14 Alleviates Premature Ovarian Failure by Inhibiting Ferroptosis Through SLC7A11/GPX4","authors":"Rong Wu, Ting Wang, Xiaofeng Xu, Ying Wang, Jingjing Hu, Wenjuan Yang, Xiao Wu, Zhaolian Wei","doi":"10.1002/ddr.70110","DOIUrl":"https://doi.org/10.1002/ddr.70110","url":null,"abstract":"<div>\u0000 \u0000 <p>Premature ovarian failure (POF) is a complex condition marked by early ovarian decline, reduced follicular reserve, and compromised oocyte quality. Oxidative stress (OS) and ferroptosis are critical drivers of POF progression. This study investigates the therapeutic potential of Phoenixin-14 (PNX-14) in alleviating POF in rats by modulating granulosa cell (GC) ferroptosis through the ATF4/SLC7A11/GPX4 signaling pathway. Cisplatin-induced POF rat models were used to evaluate PNX-14's effects on ovarian function, oxidative stress (MDA/SOD), and ferroptosis. Serum PNX-14 levels, GPR173 expression, body/ovarian weights, follicle development, and oxidative markers were analyzed. In cisplatin-induced POF rat models, serum PNX-14 levels and GPR173 expression were significantly downregulated, suggesting potential impairment of the PNX-14/GPR173 axis. PNX-14 administration improved body and ovarian weights, restored ovarian tissue structure, and reduced oxidative damage, as evidenced by reduced MDA levels and enhanced SOD activity. At the molecular level, PNX-14 suppressed ferroptosis in GCs by enhancing ATF4 expression, which in turn upregulated SLC7A11 and GPX4, critical components of cellular antioxidant defense. Caspase-3 assays suggested minimal apoptosis in cisplatin-treated cells. PNX-14 showed similar effectiveness to Fer-1 in reducing cisplatin-induced ferroptosis, evidenced by restored viability, lower Fe²⁺, and reduced MDA, without added benefit when combined. Silencing ATF4 reversed the beneficial effects of PNX-14 on GC viability and ferroptosis, confirming the pivotal role of ATF4 in mediating the protective effects of PNX-14. These results suggest that PNX-14 alleviates POF by inhibiting GC ferroptosis through the ATF4/SLC7A11/GPX4 axis, providing a potential therapeutic strategy for POF management.</p>\u0000 </div>","PeriodicalId":11291,"journal":{"name":"Drug Development Research","volume":"86 5","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-06-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144367361","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mohd Shahrukh, Shadaan Ahmad, Mohammed Zaafar, Nazeer Hasan, Farhan Jalees Ahmad
{"title":"Management of Alzheimer's Disease With Nanotechnological Interventions and Novel Therapeutics","authors":"Mohd Shahrukh, Shadaan Ahmad, Mohammed Zaafar, Nazeer Hasan, Farhan Jalees Ahmad","doi":"10.1002/ddr.70120","DOIUrl":"https://doi.org/10.1002/ddr.70120","url":null,"abstract":"<div>\u0000 \u0000 <p>Alzheimer's disease (AD) is a progressive neurodegenerative condition marked by cognitive deterioration, β-amyloid plaque buildup, intracellular tangles, and significant neuronal loss. The increasing prevalence of AD, along with its substantial economic burden, underscores the urgent need for effective therapeutic strategies in the near future. The challenge is early diagnosis and management, hindered by the lack of reliable biomarkers. Currently, there is no definitive cure for AD. Attaining improved therapeutic outcomes necessitates delivering optimal drug concentrations to the central nervous system (CNS) by effectively penetrating the blood–brain barrier (BBB). Recently, nanotechnology has emerged as a promising approach to address this challenge, enhancing brain-targeted drug delivery while highlighting recent advancements and future potential. Additionally, novel targeted therapies such as genetic therapeutics, stem cell therapy, and immunotherapy approaches overcome AD-based challenges, enhance treatment efficacy, and improve patient compliance. This review highlights recent advancements in the treatment of AD, focusing on nanotechnology-based drug delivery systems, and also explores genetic therapeutics, stem cell therapy, and immunotherapy approaches. Overall, the review provides a comprehensive overview of these therapeutic approaches, shedding light on the evolving landscape of AD treatment and the challenges that lie ahead.</p>\u0000 </div>","PeriodicalId":11291,"journal":{"name":"Drug Development Research","volume":"86 5","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-06-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144367364","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Aisha A. K. Al-Ashmawy, Mohamed Abdelraof, Asmaa Saleh, Aladdin M. Srour
{"title":"Novel Benzimidazole-Pyridine-Phenylalkanesulfonate Hybrids: Design, Synthesis, Antimicrobial Screening, Lanosterol 14α-Demethylase Inhibition Properties and in Silico Studies","authors":"Aisha A. K. Al-Ashmawy, Mohamed Abdelraof, Asmaa Saleh, Aladdin M. Srour","doi":"10.1002/ddr.70122","DOIUrl":"https://doi.org/10.1002/ddr.70122","url":null,"abstract":"<div>\u0000 \u0000 <p>A newly designed benzimidazole-pyridine-pheylalkanesulfonate hybrids (<b>3a-r</b>) were synthesized through the regioselective Michael addition reaction between 2-acetyl-1-substituted-benzimidazole (<b>1</b>) and dicyano vinyl alkanesulfonate (<b>2</b>) to evaluate their activity against diverse microbial pathogens including <i>Candida albicans</i> (a unicellular fungus), <i>Bacillus subtilis</i> and <i>Staphylococcus aureus</i> (Gram-positive bacteria), as well as <i>Pseudomonas aeruginosa</i>, <i>Klebsiella pneumoniae</i>, and <i>Salmonella typhimurium</i> (Gram-negative bacteria). Although most of the tested hybrids showed promising antimicrobial potentiality, the MIC was calculated for the two precursors <b>1a</b>,<b>b</b> in addition to derivatives <b>3b</b>, <b>3 g</b>, <b>3k</b>, <b>3 m</b> and <b>3p</b> that found to prevent the proliferation of all microbial pathogens with different ratios in comparison to the reference drug used. Compound <b>3k</b> demonstrated the best anti-candida properties with MIC = 5 <i>μ</i>g/mL (amphotericin B, MIC = 20 <i>μ</i>g/mL), and it was of equal efficacy to ciprofloxacin against <i>Staphylococcus aureus</i> with MIC = 20 <i>μ</i>g/mL. Derivatives <b>3b</b> and <b>3 m</b> displayed equipotent activity to ciprofloxacin with MICs = 10 <i>μ</i>g/mL against <i>S. typhimurium</i> and <i>B. subtilis</i>, respectively. Moreover, derivative <b>3 m</b> exhibited 42.1% biofilm inhibition against <i>P. aeruginosa</i> (compared with 39.8% for Ciprofloxacin). An acceptable safety profile of the most potent derivatives was detected. The impact of the <b>3b</b> and <b>3k</b> hybrids on the tested microbial pathogens was visualized using CLSM. Furthermore, <b>3k</b> inhibited the Lanosterol 14α-demethylase (CYP51) with IC<sub>50</sub> = 4.2 <i>μ</i>M (fluconazole IC<sub>50</sub> = 0.6 <i>μ</i>M). Furthermore, in silico studies including ADME prediction for the five promising hits and molecular docking simulation of <b>3k</b> in the demethylase binding site were performed.</p>\u0000 </div>","PeriodicalId":11291,"journal":{"name":"Drug Development Research","volume":"86 5","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-06-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144367488","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kun Du, Yanan he, Jinyang Fu, Guimin Xue, Zhiqiang Zhang, Xiaokun Li, Yanle Zhi
{"title":"Design, Synthesis, and Biological Evaluation of Potent FLT3 Inhibitor for Acute Myeloid Leukemia (AML) Treatment","authors":"Kun Du, Yanan he, Jinyang Fu, Guimin Xue, Zhiqiang Zhang, Xiaokun Li, Yanle Zhi","doi":"10.1002/ddr.70119","DOIUrl":"https://doi.org/10.1002/ddr.70119","url":null,"abstract":"<div>\u0000 \u0000 <p>Acute myeloid leukemia (AML) is a clonal malignant proliferative disease of myeloid progenitor cells in the hematopoietic system, with a lower than 5-year overall survival rate. At present, three FLT3 inhibitors have been approved, but these drugs are prone to cause resistance after a period of medication. Developing new FLT3 inhibitors with novel structures is an effective strategy to enhance drug treatment efficacy. This study presents an extension of our effort to design and synthesize a series of novel pyrimidine-2,4-diamine derivatives as inhibitors of FLT3. The most active compound, <b>7r</b>, showed significant inhibition against FLT3 with IC<sub>50</sub> value of 7.82 nM. In addition, <b>7r</b> exhibited prominent anticancer activities against AML cell lines, such as MV4-11 (IC<sub>50</sub> = 46.07 nM) and MOLM-13 (IC<sub>50</sub> = 51.6 nM). Compound <b>7r</b> inhibited phosphorylation of FLT3 pathways in a dose-dependent manner in MV4-11 cell lines.</p></div>","PeriodicalId":11291,"journal":{"name":"Drug Development Research","volume":"86 5","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144315279","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Advancements in Organoid-Based Drug Discovery: Revolutionizing Precision Medicine and Pharmacology","authors":"Dilpreet Singh, Akshay Thakur, Rakesh, Akshay kumar","doi":"10.1002/ddr.70121","DOIUrl":"https://doi.org/10.1002/ddr.70121","url":null,"abstract":"<div>\u0000 \u0000 <p>Organoids, 3D cellular models derived from stem cells, have revolutionized drug testing by providing human-relevant systems for modeling diseases and testing drug efficacy. Unlike traditional 2D cell cultures or animal models, organoids closely resemble the complex architecture and function of human tissues, offering more accurate predictions of drug responses. Researchers are increasingly utilizing these models in oncology, neurology, liver toxicity, and personalized medicine. Recent advances in gene editing (e.g., CRISPR-Cas9), multi-omics technologies, and organoid-on-chip systems have further enhanced the capabilities of organoids in drug discovery. CRISPR-Cas9 allows for precise modeling of genetic disorders, while multi-omics approaches integrate transcriptomics, proteomics, and metabolomics to provide deeper insights into drug metabolism and toxicity. Organoid-on-chip platforms combine organoid culture with microfluidic systems, enabling the simulation of organ interactions and real-time drug testing. AI and machine learning models now enhance these platforms by predicting drug responses and optimizing high-throughput screening. Despite these advancements, challenges such as scalability, reproducibility, and the incomplete recapitulation of complex organ functions remain. Organoids hold the promise of significantly reducing reliance on animal models, improving the accuracy of drug testing, and paving the way for personalized treatments. This review outlines the latest innovations in organoid-based drug discovery, highlighting their potential to transform modern pharmacology and precision medicine, while acknowledging the ongoing efforts to address existing limitations.</p>\u0000 </div>","PeriodicalId":11291,"journal":{"name":"Drug Development Research","volume":"86 4","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144291906","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ahmed A. Allam, Hassan A. Rudayni, Noha A. Ahmed, Faris F. Aba Alkhayl, Al Mokhtar Lamsabhi, Emadeldin M. Kamel
{"title":"β-Glucuronidase Inhibition in Drug Development: Emerging Strategies for Mitigating Drug-Induced Toxicity and Enhancing Therapeutic Outcomes","authors":"Ahmed A. Allam, Hassan A. Rudayni, Noha A. Ahmed, Faris F. Aba Alkhayl, Al Mokhtar Lamsabhi, Emadeldin M. Kamel","doi":"10.1002/ddr.70118","DOIUrl":"https://doi.org/10.1002/ddr.70118","url":null,"abstract":"<div>\u0000 \u0000 <p>β-glucuronidase (βG) is a critical enzyme involved in the hydrolysis of glucuronide conjugates, significantly influencing drug metabolism, detoxification processes, and enterohepatic circulation. Although essential for maintaining physiological homeostasis, dysregulated βG activity has been implicated in diverse pathological conditions, including drug-induced toxicity, inflammation, and hormone-dependent cancers. Specifically, microbial βG expressed by gut microbiota can reactivate glucuronide-conjugated drugs, leading to adverse reactions through increased drug toxicity and reduced therapeutic efficacy. Consequently, inhibition of βG has emerged as an attractive therapeutic approach to reduce chemotherapy-induced toxicity, gastrointestinal complications, and metabolic disorders. This review systematically examines recent progress in the discovery, characterization, and optimization of βG inhibitors, focusing on natural products, synthetic molecules, and microbiome-targeted agents. Structure–activity relationship analyses reveal crucial functional groups and chemical modifications necessary for enhancing inhibitor potency, selectivity, and bioavailability. In addition, contemporary advances in βG inhibitor evaluation through enzyme kinetics, molecular docking simulations, high-throughput screening, and preclinical animal models are discussed, alongside essential pharmacokinetic parameters, including absorption, distribution, metabolism, excretion, and potential drug-drug interactions. Furthermore, emerging approaches such as microbiome modulation, CRISPR-based enzyme engineering, and combination therapies are explored. Despite promising preclinical outcomes, significant challenges remain regarding clinical translation, such as selectivity, bioavailability, and regulatory compliance. Ultimately, this review highlights future opportunities in precision medicine, emphasizing personalized βG inhibitor development to optimize therapeutic safety and effectiveness across various disease states.</p>\u0000 </div>","PeriodicalId":11291,"journal":{"name":"Drug Development Research","volume":"86 4","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144291847","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sukanya Sukanya, Aina Bellver-Sanchis, Bhanwar Singh Choudhary, Sunil Kumar, Belén Pérez, Antón Leandro Martínez Rodríguez, Jose Brea, Carmen Escolano, Christian Griñán-Ferré, Ruchi Malik
{"title":"Exploring 2-Thioxo-1,2,3,4-tetrahydropyrimidines as Dual Acting GSK-3β/Aβ Aggregation Inhibitors: Implications for Alzheimer's Disease Treatment","authors":"Sukanya Sukanya, Aina Bellver-Sanchis, Bhanwar Singh Choudhary, Sunil Kumar, Belén Pérez, Antón Leandro Martínez Rodríguez, Jose Brea, Carmen Escolano, Christian Griñán-Ferré, Ruchi Malik","doi":"10.1002/ddr.70112","DOIUrl":"https://doi.org/10.1002/ddr.70112","url":null,"abstract":"<div>\u0000 \u0000 <p>The etiology of Alzheimer's disease (AD) is complex and multifactorial. There is a pressing need for therapies that can prevent or slow AD progression. Consequently, drug development has shifted from single-target approaches to multi-faceted strategies that emphasize early intervention rather than late-stage treatment. One promising target is glycogen synthase kinase-3β (GSK-3β), an enzyme implicated in tau hyperphosphorylation and Aβ plaque formation. Based on our earlier work, we synthesized 25 2-thioxo-1,2,3,4-tetrahydropyrimidine derivatives designed as GSK-3β inhibitors, tau phosphorylation inhibitors, and Aβ accumulation. Two compounds emerged as particularly effective: compound 63 (IC<sub>50</sub> = 1.69 µM) and compound 66 (IC<sub>50</sub> = 0.90 µM), with compound 66 identified as an ATP-competitive inhibitor of GSK-3β. Further pharmacokinetic studies and in vitro drug metabolism assessments were conducted, followed by in vivo efficacy studies using <i>Caenorhabditis elegans</i>. Notably, these compounds reduced phosphorylated tau levels in the BR5706 strain and decreased Aβ aggregate deposition in the CL2006 strain. Molecular Dynamic (MD) simulations were also performed on both compounds. These findings provide valuable insights into GSK-3β drug development and highlight the potential of these inhibitors as therapeutic candidates for AD by targeting both tau and Aβ, the two pathological hallmarks of AD.</p>\u0000 </div>","PeriodicalId":11291,"journal":{"name":"Drug Development Research","volume":"86 4","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144291905","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Clinical Investigation of Tyrosinase Inhibitors: Past, Present, and Future","authors":"Yuanyuan Wang, Ruijia Jiang, Baichen Xiong, Jiawei Zhu, Jingjing Sang, Hongtao Li, Chen Chen, Ziwei Xu, Weiting Zhang, Yao Chen, Feng Feng, Haopeng Sun","doi":"10.1002/ddr.70113","DOIUrl":"https://doi.org/10.1002/ddr.70113","url":null,"abstract":"<div>\u0000 \u0000 <p>Tyrosinase (EC 1.14.18.1) is a pivotal enzyme that catalyzes the conversion of L-tyrosine to dopaquinone through a dual oxidation process, initiating melanin biosynthesis. Melanin plays a critical role in various biological processes, and its overproduction is associated with multiple conditions. Tyrosinase plays a crucial role in immune regulation by regulating the activity of immune cells and enhancing the immune response of the body. It is essential for maintaining skin health and preventing autoimmune diseases. In addition, tyrosinase has shown potential in immunotherapy, especially in the treatment of malignant melanoma and autoimmune diseases such as vitiligo. Inhibiting tyrosinase to reduce melanin synthesis has emerged as a promising therapeutic strategy with applications in skin whitening, melasma treatment, acne management, Parkinson's disease (PD) intervention, melanoma prevention, and overcoming immunotherapy resistance. By leveraging the tyrosinase-related comprehensive data documented in the BRENDA database, we have systematically summarized the effective information, including its classification, structural characteristics, catalytic functions, biosynthesis pathways, substrate specificity profiles, reaction products, and associated disease mechanisms, and so forth. This review comprehensively examines the therapeutic mechanisms, development history, and current clinical status of tyrosinase inhibitors at preclinical and advanced stages. We highlight recent research progress, focusing on evidence from animal models, preclinical studies, and human clinical trials across different indications. Additionally, we critically analyze the challenges and limitations in the field and provide insights into future directions for optimizing tyrosinase inhibitors. By synthesizing current knowledge and advancements, this review aims to underscore the therapeutic potential of tyrosinase inhibition and its role in addressing diverse medical needs.</p></div>","PeriodicalId":11291,"journal":{"name":"Drug Development Research","volume":"86 4","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-06-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144291898","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Kaempferitrin Regulates the Proliferation, Metastasis, and Immune Escape of Nonsmall Cell Lung Cancer by Inhibiting the Akt/NF-κB Pathway","authors":"Zhenliang Shi, Yimeng Shen, Xin Liu, Sipei Zhang","doi":"10.1002/ddr.70117","DOIUrl":"https://doi.org/10.1002/ddr.70117","url":null,"abstract":"<div>\u0000 \u0000 <p>Nonsmall cell lung cancer (NSCLC) is the most popular type of lung cancer with high morbidity. Kaempferitrin possesses the antitumor effect, while its role in NSCLC development and metastasis remains elusive. Thus, this study aimed to clarify the influence of Kaempferitrin on NSCLC development and elucidate the possible mechanism. In this study, cell viability, apoptosis, invasion, and angiogenesis were assessed using CCK-8, flow cytometry, Transwell invasion, and tube formation assays, respectively. The NF-κB nuclear translocation was observed via Immunofluorescence. In vivo, tumor growth was monitored in a xenograft model, with HE and TUNEL staining assessing tissue damage and apoptosis. Results revealed that Kaempferirin decreased NSCLC cell viability, induced cell apoptosis and suppressed cell motility dose-dependently. Additionally, Kaempferirin restrained the immune escape of NSCLC cells and inactivated the AKT/NF-κB pathway. SC79 could counteract the above effects of Kaempferirin on A549 cells. Moreover, Kaempferirin restrained tumor growth, metastasis and immune escape of NSCLC In vivo. In summary, Kaempferirin inhibited cell proliferation, invasion, angiogenesis, and immune escape and promoted apoptosis of NSCLC by inactivating the AKT/NF-κB pathway. Kaempferirin may be a hopeful drug for NSCLC therapy.</p>\u0000 </div>","PeriodicalId":11291,"journal":{"name":"Drug Development Research","volume":"86 4","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-06-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144264368","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Novel Metronidazole Conjugates as Antimicrobial Agents","authors":"Erol Akgün, Melike Demirayak, Leyla Yurttaş, Ülkiye Dudu Gül, Şeref Demirayak","doi":"10.1002/ddr.70114","DOIUrl":"https://doi.org/10.1002/ddr.70114","url":null,"abstract":"<p>Metronidazole (MTZ) is one of the oldest and still used anti-infective nitroimidazole group drug. Although it is effective against anaerobic bacteria, protozoa, and parasites in clinical settings, it lacks efficacy against aerobic microorganisms. Due to its efficient molecular structure and synthetic usability due to the alcohol group in its framework, medicinal chemists aimed to reach new more effective molecules such as MTZ-hybrids. In this study, 2-[(benzimidazole/benzoxazole/benzothiazol-2-yl)thio]-<i>N</i>-[2-(2-methyl-5-nitro-1<i>H</i>-imidazol-1-yl)ethyl]acetamide (<b>5a−5j</b>) derivatives were synthesized and their antimicrobial and antifungal effects on aerobic bacteria and <i>Candida</i> spp. were investigated. Notably, most of newly designed conjugates displayed higher potency than MTZ itself, especially against Gram-positive strains. Furthermore, chlorinated heterocyclic moieties provided the strongest effects. Docking studies using <i>E. coli</i> nitroreductase (PDB: 1IDT) revealed potential interactions with the flavin mononucleotide (FMN) cofactor, suggesting that these hybrids may undergo nitro-group reduction analogous to MTZ. Additionally, pharmacokinetic predictions indicated generally favorable profiles.</p>","PeriodicalId":11291,"journal":{"name":"Drug Development Research","volume":"86 4","pages":""},"PeriodicalIF":3.5,"publicationDate":"2025-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/ddr.70114","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144245067","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}