EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY

Synthesis of novel deuterated EGFR/ALK dual-target inhibitors and their activity against non-small cell lung cancer
Nie H, Zhang S, Wang L, Wang M, Qiu J, Jia F, Li X, Tian G and An B
EGFR and ALK are common driver genes in NSCLC, and more patients with these mutations are being identified due to medical advances. Thus, developing dual-target EGFR/ALK inhibitors is crucial. In this study, 10 novel small molecules were designed and synthesized. CCK8 experiments revealed that compound (-)-9a exhibited the best anti-tumor activity, with IC values of 1.08 ± 0.07 nM for EGFR and 2.395 ± 0.023 nM for ALK mutant tumor cells. Studies show that compound (-)-9a can inhibit phosphorylated proteins in EGFR, ALK, and BRK signaling pathways and halt the cell cycle, leading to reduced mitochondrial membrane potential and apoptosis in tumor cells. Additionally, (-)-9a not only directly targets tumor cells but also exhibits potential immune-enhancing effects. Furthermore, evaluations conducted in animal models have demonstrated that this drug effectively reduces tumor growth in vivo. In summary, (-)-9a boasts dual-targeting, potent antitumor activity, and immune-enhancing potential, presenting vast potential as a next-gen anticancer drug.
Design, synthesis and biological evaluation of novel β-carbolines as antitumor agents via targeting autophagy in colorectal cancer
Ao J, Lai C, Wu X, Chen Z, Yang W, Qiu L, Li X and Cao R
A series of novel β-carbolines with a flexible amino side chain at positions 1 and 3, respectively, were designed, synthesized and evaluated as potential antitumor agents. The results revealed that most of the compounds exhibited a broad spectrum of antiproliferative activity with IC value lower than 20 μM against human tumor cell lines. Among them, compound 2f was the most potent antiproliferative agent with IC value below 5.0 μM against human tumor cell lines. Subsequent studies on the in vivo antitumor efficacy of the representative compound 2f demonstrated its ability to hinder tumor progression and significantly diminish tumor mass in a mouse model of colorectal cancer. Further investigation on mechanisms of action showed that compound 2f induced autophagy via the ATG5/ATG7 pathway in HCT116 cells. These compounds may contribute to the development of therapeutic agents for colorectal cancer.
Design and synthesis of novel benzoic acid derivatives as striatal-enriched protein tyrosine phosphatase (STEP) inhibitors with neuroprotective properties
Jiang C, Liu R, Chang Y, Zhang S, Li X, Zhao Z, Quan M, Wang Q, Zhou H, Hou X and Fang H
As a central nervous system-specific member of the protein tyrosine phosphatase (PTP) family, the striatal-enriched protein tyrosine phosphatase (STEP) is an attractive drug target for neurodegenerative diseases. Here, we reported the discovery of a series of benzoic acid derivatives as new STEP inhibitors. Among them, compound 14b exhibited good STEP inhibitory activity and displayed selectivity against other PTPs. The neuroprotective activity of compound 14b was evaluated against glutamate-induced oxidative cell death in HT22 cells. Results indicated that compound 14b co-treatment prevented cell death and reduced cellular ROS accumulation. Compound 14b inhibited cell apoptosis by upregulating BCL-2 expression and downregulating BAX and C-caspase3 expression. Moreover, compound 14b was also found to provide neuroprotection to primary cortical neurons after oxygen-glucose deprivation/reoxygenation (OGD/R). Further structural elaboration of compound 14b may provide new drug candidates for neurodegenerative diseases.
Efficacy and molecular mechanisms of hesperidin in mitigating Alzheimer's disease: A systematic review
Han X, Zhang Y, Zhang L, Zhuang Y and Wang Y
Hesperidin, a flavonoid glycoside, is a natural phenolic compound that has broad biological effects. Increasing evidence suggests that hesperidin inhibits the occurrence and development of neurodegenerative diseases, including Alzheimer's disease (AD). This article reviews the neuropharmacological mechanisms of hesperidin in the prevention and treatment of AD through in vitro and in vivo studies. A systematic review of preclinical studies was conducted using PubMed, Web of Science, Scopus, and Google Scholar (up to July 1, 2024). The neuroprotective potential of hesperidin was mediated through mechanisms such as inhibition of β-amyloid (Aβ) aggregation, enhancement of endogenous antioxidant defense functions, reduction of neuroinflammation and apoptosis, improvement of mitochondrial dysfunction, regulation of autophagy, and promotion of neurogenesis. Despite various preclinical studies on the role of hesperidin in AD, its exact effects on humans remain unclear. Few clinical trials have indicated that dietary supplements rich in hesperidin can improve cerebral blood flow, cognition, and memory performance. The neuroprotective effect of hesperidin may be exerted via regulating different molecular pathways, including the RAGE/NF-κB, Akt/Nrf2, and AMPK/BDNF/CREB pathways. However, further clinical trials are needed to confirm the neuroprotective effects of this natural flavonoid compound and to assess its safety.
Targeting ferroptosis offers therapy choice in sepsis-associated acute lung injury
Wang Y, Wang W, Zhang Y, Fleishman JS and Wang H
Sepsis-associated acute lung injury (SALI) is a common complication of sepsis, consisting of a dysfunctional host response to infection-mediated heterogenous complexes. SALI is reported in up to 50 % of patients with sepsis and causes poor outcomes. Despite high incidence, there is a lack of understanding in its pathogenesis and optimal treatment. A better understanding of the molecular mechanisms underlying SALI may help produce better therapeutics. The effects of altered cell-death mechanisms, such as non-apoptotic regulated cell death (RCD) (i.e., ferroptosis), on the development of SALI are beginning to be discovered, while targeting ferroptosis as a meaningful target in SALI is increasingly being recognized. Here, we outline how a susceptible lung alveoli may develop SALI. Then we discuss the general mechanisms underlying ferroptosis, and how it contributes to SALI. We then outline the chemical structures of the emerging agents or compounds that can protect against SALI by inhibiting ferroptosis, summarizing their potential pharmacological effects. Finally, we highlight key limitations and possible strategies to overcome them. This review suggests that a detailed mechanistic and biological understanding of ferroptosis can foster the development of pharmacological antagonists in the treatment of SALI.
High potency 3-carboxy-2-methylbenzofuran pendrin inhibitors as novel diuretics
Master RJ, Karmakar J, Haggie PM, Anthony-Tan J, Chu T, Verkman AS, Anderson MO and Cil O
Pendrin (SLC26A4) is an anion exchanger expressed in epithelial cells of kidney and lung. Pendrin inhibition is a potential treatment approach for edema, hypertension and inflammatory lung diseases. We have previously identified first-in-class pendrin inhibitors by high-throughput screening, albeit with low potency for pendrin inhibition (IC ∼10 μM). Here, we performed a de novo small molecule screen with follow-on structure-activity studies to identify more potent pendrin inhibitors. Screening of 50,000 synthetic small molecules identified four novel classes of pendrin inhibitors with diverse scaffolds, including 5-benzyloxy-2-methylbenzofurans, N-aryl urea substituted 5-methyltryptamines, N-aryl urea substituted anthranilic acids, and substituted N-benzyl 3-carboxyindoles. The most potent inhibitor from the initial screen, a 3-carboxy-2-methylbenzofuran (1a), had IC of 4.1 μM. Structure-activity studies using 732 benzofuran analogs identified 1d with IC ∼ 0.5 μM for pendrin inhibition. Selectivity studies showed that 1d has minimal or no activity against related ion channels/transporters including SLC26A3, SLC26A6 and CFTR at high concentrations. 1d administration to mice at 10 mg/kg had no effect on urine volume when used alone, but potentiated the diuretic effect of furosemide by 45 %. In conclusion, we have identified novel pendrin inhibitors with greatly improved potency and good in vivo efficacy. These compounds can be used as pharmacological tools to study the roles of pendrin, and potentially developed as drug candidates for edema, hypertension and lung diseases.
Expanding the therapeutic window of gramicidin S towards a safe and effective systemic treatment of methicillin-resistant S. aureus infections
Wang Y, Kalyvas JT, Evans JD, Toronjo-Urquiza L, Horsley JR and Abell AD
The rise of multidrug-resistant bacteria, such as Methicillin-resistant Staphylococcus aureus (MRSA), necessitates the development of new antibacterial therapies. Antimicrobial peptides offer a promising alternative to conventional antibiotics due to their unique mechanisms of action. Gramicidin S exhibits potent bactericidal activity against S. aureus, however, high haemolytic toxicity currently limits its application to topical use. A new series of gramicidin S analogues is presented with rational modifications to the β-turn and β-strand regions, to reduce haemolytic and nephrotoxic effects, while preserving antibacterial potency. The minimum inhibitory concentration (MIC) for each analogue was determined against benchmark methicillin-sensitive S. aureus (MSSA) and MRSA clinical isolates, with toxicity characterised in vitro using human red blood cells and human embryonic kidney cells (HEK-293). Peptide 12 demonstrated a significant two-fold increase in antibacterial activity against both MSSA and MRSA (MIC: 2 μg/mL) compared to gramicidin S (MIC: 4 μg/mL), albeit with increased cytotoxicity. Similarly, peptide 15 showed exceptional efficacy (MIC: 3 μg/mL), but with reduced cytotoxicity, culminating in a two-fold improvement to the therapeutic index (TI) of gramicidin S. Peptides 14 (HC: 50.48 ± 1.15 μg/mL, IC: 38.09 μg/mL) and 16 (HC: 84.09 ± 1.02 μg/mL, IC: 12.60 μg/mL) also significantly reduced haemolytic toxicity and nephrotoxicity, compared to gramicidin S (HC: 12.34 ± 0.27 μg/mL, IC: 6.45 μg/mL). Detailed NMR, CD and computational modelling were used to provide critical insights into how molecular conformation influences both antibacterial potency and cytotoxicity. Collectively, these results expand the therapeutic window of gramicidin S by up to 12-fold, with negligible cytotoxicity observed at concentrations well beyond the acceptable safety threshold, which indicates the potential for safe systemic administration in the treatment of infection caused by resistant pathogens.
Designed and synthesized novel tripeptides targeting diabetes and its related pathologies
Khan M, Khan S, Ahmad S, Alshammary FL, Mahmood T, Khan MS and Rahim M
In diabetes and its associated pathologies, glycation, α-amylase, and α-glucosidase play crucial roles. This study introduces a novel tripeptide, RWW, designed to target glycation and key enzymes in diabetes management. Using in silico methods, RWW was optimized to interact with the glycation-prone Human serum albumin (HSA) sites, as well as inhibit α-amylase and α-glucosidase. Molecular docking and dynamics confirmed its stability. In-vitro assays confirmed RWW's potent inhibition of glycation (84.00 %) and enzyme activities, while in-vivo experiments demonstrated its hypoglycemic and lipid-lowering effects in diabetic mice. Histopathological analysis of kidney tissues further highlighted its protective impact. RWW represents a promising anti-diabetic candidate with dual therapeutic functions.
Discovery of novel biaryl urea derivatives against IL-1β release with low toxicity based on NEK7 inhibitor
Wang L, Zhu K, Tian Z, Wang H, Jia Y, Feng C, Qi L, Tang W and Hu Y
Aberrant activation of NLRP3 inflammasome is involved in various inflammatory diseases, making it a promising target for therapeutic intervention. NEK7, a member of the NIMA-related kinase (NEK) family, functions as a key NLRP3-binding protein and plays a crucial role in the regulation of NLRP3 inflammasome assembly and activation. Thus, disrupting NLRP3-NEK7 interactions by targeting NEK7 could be a promising strategy to inhibit the activation of NLRP3 inflammasome. In this work, a series of novel urea derivatives were designed and synthesized based on the reported NEK7 inhibitors. Among these, compound 23 exhibited potent activity against IL-1β release with low cytotoxicity. Moreover, compound 23 enhanced the thermal stability of NEK7 and disrupted the NLRP3-NEK7 interaction, thereby regulating NLRP3 inflammasome assembly.
Hypoxia-selective prodrug restrains tumor cells through triggering mitophagy and inducing apoptosis
Wang F, Song L, Xu Q, Jia A, Meng X, Jiang H and Zhang R
Hypoxia is a common feature of various solid tumors, which reduces the sensitivity of tumor cells to both radiotherapy and chemotherapy. However, hypoxia also presents an opportunity for tumor-selective therapy. The prodrug strategy, leveraging the hypoxic nature of the tumor microenvironment, shows significant potential for clinical application. Here we present CHD-1, a hypoxia-activated antitumor prodrug that activates in hypoxic environments, effectively inhibiting hypoxic tumor cells while exhibiting no toxicity to normoxic cells. CHD-1 impairs mitochondrial morphology and membrane potential of hypoxic tumor cells, further triggers excessive mitophagy and induces apoptosis. Moreover, prodrug CHD-1 significantly inhibits HeLa xenograft growth in vivo, and shows lower toxicity than parent molecule in an acute toxicity assessment in animal models. This study introduces a promising hypoxia-activated antitumor prodrug with strong potential for further development in hypoxic tumor therapy.
Navigating cancer therapy: Harnessing the power of peptide-drug conjugates as precision delivery vehicles
Sagar B, Gupta S, Verma SK, Reddy YVM and Shukla S
Cancer treatment is a formidable challenge due to the adverse effects associated with non-selective therapies like chemotherapy and radiotherapy. This review article primarily centers on the application of Peptide-Drug Conjugates (PDCs) for delivering cancer treatment. PDCs represent a promising class of precision medicines, harnessing the unique attributes of peptides in conjunction with non-peptide components. The covalent linking of peptides and drugs through specialized connectors characterizes PDCs. These constructs play a pivotal role in delivering drugs directly to tumor sites with high precision. PDCs encompass three pivotal components: a targeting ligand, a cytotoxic ligand, and a carefully chosen linker. The selection of these elements is crucial to maximize the efficiency of PDCs. PDCs offer a multitude of advantages over conventional drug molecules, including enhanced specificity, reduced off-target effects, and an improved therapeutic profile. The peptide component within PDCs can be customized to specifically adhere to disease-specific receptors or biomarkers, facilitating targeted drug delivery and accumulation in afflicted cells or tissues. This targeted approach enables the controlled release of therapeutic payloads at the localized site, resulting in heightened effectiveness and minimized systemic toxicity. Diverse linker strategies are employed to ensure the stable connection between the peptide and non-peptide components, ensuring controlled drug release at the desired location of action. The peptides utilized in these treatments encompass cell-penetrating peptides, peptides designed to target tumor cells, and those aimed at the nucleus of cancer cells. While certain clinical trials have been conducted, and some PDCs are currently in use for cancer treatment, it's essential to acknowledge that PDCs have their limitations, such as low stability in plasma, fast elimination and limited oral bioavailability. Ongoing research endeavors seek to surmount these challenges and further establish PDCs as potent agents for cancer treatment. This review sheds light on recent advancements in the design, delivery, and applications of PDCs, while also highlighting the prevailing challenges and charting a path for future research directions.
Design, synthesis and investigation of biological activity and mechanism of fluoroaryl-substituted derivatives at the FL118 position 7
Wang W, Wang R, An L, Li L, Xiong H, Li D, Dong F, Lei J, Wang M, Yang Z, Wang H, Ling X, Fountzilas C, Li F and Li Q
Addition of fluorine atoms into chemical compounds is a validated strategy to enhance their physical, chemical and biological properties. In this study, FL118, a novel camptothecin-related small molecule known for its unique mechanism of action and superior antitumor efficacy, was utilized as a foundational drug platform. By replacing the hydrogen atom at position 7 of FL118 with a fluoroaryl group, a diverse array of FL118 derivatives were synthesized. Our investigations revealed that the majority of these newly synthesized compounds exhibited improved cytotoxicity compared to FL118, with some demonstrating enhanced in vivo antitumor efficacy. Among these derivatives, compound 7h stood out and was subjected to detailed analysis. Compound 7h demonstrated a remarkable ability to inhibit colorectal cancer (CRC) cell colony formation and cell migration, while also promoting reactive oxygen species (ROS) production and CRC cell apoptosis. Notably, our studies unveiled that the presence of DDX5 could modulate Topoisomerase I (Top1) activity, a process effectively reversed by a low concentration of 7h, but not SN38. Moreover, only 7h was able to decrease DDX5 expression, SN38 was not. Molecular docking studies further supported the binding of 7h to DDX5. Interestingly, although both 7h and SN38 exhibited similar inhibitory effects on Top1 activity, only 7h, and not SN38, could inhibit DDX5. These findings not only pave the way for deeper mechanistic explorations of FL118 and its derivatives in cancer research but also position the identified compound 7h as a promising candidate for further development.
Structure-activity relationship study of novel evodiamine amino acid conjugates with potent anti-colorectal cancer efficacy
Chen S, Zhang X, Mo H, Peng Y, An Z, Wu J, Wei X, Zhang S, Xiong Y, Jiang W, Peng X, Zhuo L, Lei Z, Wang Z and Hu Z
Evodiamine has been a promising lead structure with broad-spectrum antitumor activity. Druggability optimization is the most challenging part of evodiamine-based lead-to-candidate campaign. Amino acids as building blocks for conjugates are widely incorporated into approved drug and drug candidates, demonstrating highly attractive druggability. Herein, a series of evodiamine amino acid conjugates were designed and synthesized based on the evodiamine lead compound (±)-8b discovered in our previous work. The structure-activity relationship (SAR) studies culminated in the identification of a promising conjugate (-)-15h featuring a N-Boc-l-glutamine group and a chiral carbon atom (sinister), which exhibited nanomolar antiproliferative activity against LoVo and RKO colorectal cancer cells. Moreover, (-)-15h could inhibit topoisomerases I, arrest the cell cycle in the G2/M phase, and induce apoptosis. Importantly, (-)-15h (tumor growth inhibition rate was 82.53 % in 40 mpk) showed better efficacy and tolerability to that of parent compound (-)-8b (tumor growth inhibition rate was 51.22 % in 40 mpk) in LoVo xenograft model. Further, (-)-15h (tumor growth inhibition rate was 70.09 % in 40 mpk) showed comparable efficacy and better tolerability to that of topotecan (tumor growth inhibition rate was 70.67 % in 0.5 mpk) in HT-29 xenograft model. Collectively, this study further provided a strong scientific basis for amino acid-based structural modifications and a drug lead for anti-colorectal cancer applications.
Design, synthesis, and biological evaluations of 5-aryl-pyrazole-3-carboxamide derivatives as selective CB2 receptor agonists for the treatment of colitis
Jiang BE, He Y, Chen J, Jiang XW, Qiu ZL, Liang QW, Gao XL, Zhang HK, Tian HG, Liu MY, Lu WQ and Yu LF
Synthetic CB2 receptor agonists exhibit great potential in the treatment of neurodegenerative diseases, chronic and neuropathic pain, cancer, and inflammation-associated pathologies while avoiding adverse psychoactive effects caused by interactions with CB1 receptors. Herein, a class of 5-aryl-pyrazole-3-carboxamide derivatives was thus designed, synthesized, and biologically evaluated. Among the compounds tested, compound 33, one of the most potent leads, showed a remarkably high potency and selectivity at the CB2 receptor (EC = 16.2 nM, EC > 10 nM). Furthermore, 33 treatment significantly attenuate colon inflammation in a dextran sodium sulfate (DSS)-induced mouse model of colitis, supporting that CB2 receptor agonists might serve as potential therapeutics for treating colitis.
Small molecule inhibitors targeting PD-L1, CTLA4, VISTA, TIM-3, and LAG3 for cancer immunotherapy (2020-2024)
Cheng B, Lv J, Xiao Y, Song C, Chen J and Shao C
Cancer immunotherapy, leveraging antibodies, excels in targeting efficacy but faces hurdles in tissue penetration, oral delivery, and prolonged half-life, with costly production and risk of adverse immunogenic effects. In contrast, small molecule immuno-oncology agents provide favorable pharmacokinetic properties and benign toxicity profiles. These agents are well-positioned to address the limitations of antibody-based immunotherapies, augment existing treatment modalities, and achieve synergistic effects when combined with antibodies. This review, for the first time, summarizes the recent advances (2020-2024) in small molecule inhibitors targeting PD-1/PD-L1, CTLA4, VISTA, TIM-3, and LAG3, highlighting rational design, benefits, and potential limitations. It also outlines the prospects for small-molecule immunotherapy.
LAT1 transporter as a target for breast cancer diagnosis and therapy
Zhou Z, Zhu T, Zheng W, Zou Z, Shan Q, Chen Q, Wang G and Wang Y
Breast cancer is the main cause of female malignant tumor death in China. Numerous cellular molecules are associated with the onset and progression of breast cancer. However, these molecules have proven ineffective for the diagnosis and treatment of the disease, indicating a need for the identification of new biomarkers. LAT1 (SLC7A5) plays a crucial role in mediating the uptake of amino acids into breast cancer cells, influencing proliferation, invasion, migration, drug resistance, and prognosis through the mTOR signaling pathway. Notably, LAT1 exhibits differential expression across various types of breast cancer, positioning it as a promising candidate for diagnostic and therapeutic applications. Recent advancements in LAT1-targeting strategies for breast cancer have been made, particularly with the rapid developments in small molecular inhibitors and nanotechnology. In this article, we review the structure and function of LAT1, its relationship with breast cancer, and LAT1-mediated diagnostic and treatment strategies. This article specifically focuses on the LAT1-targeting strategy in breast tumors, aiming to evaluate its potential role as a novel biomarker for diagnosis and treatment.
Circumventing Imatinib resistance in CML: Novel Telmisartan-based cell death modulators with improved activity and stability
Gebhart M, Alilou M, Gust R and Salcher S
Drug resistance presents a significant challenge in cancer therapy, which has led to intensive research in resistance mechanisms and new therapeutic strategies. In chronic myeloid leukemia (CML), the introduction of Imatinib, the first tyrosine kinase inhibitor (TKI), drastically changed the outcome for patients. However, complete remission still cannot be achieved in a large number of patients in the long term. Therefore, there is a great interest in the design of new drugs to target TKI-resistant cancer cells. A promising approach to enhance the efficacy of Imatinib is the simultaneous application of cell death modulators derived from the Angiotensin II type 1 receptor blocker Telmisartan. The methyl ester (3a) of 4'-((2-propyl-1H-benzo[d]imidazol-1-yl)methyl)-[1,1'-biphenyl]-2-carboxylic acid (LEAD-acid (4)), which is the structural core of Telmisartan, has already been shown to abolish the resistance of Imatinib in TKI-insensitive CML cells at a concentration of 5 μM. As the ester was expected to be unstable in a biological environment, this study attempted to increase the stability through structural modifications. The methyl group was exchanged for longer (3b (ethyl), 3c (propyl), 3d (butyl) and branched (3e (isopropyl), 3f (tert-butyl)) alkyl chains as well as a phenyl (3g) and 4-phenoxyphenyl (3h) group. Furthermore, the esters were bioisosterically replaced with a respective substituted carboxamide (5a-h). The LEAD-amides (5a-h) showed high stability against esterases, while amidases cleaved only the carboxamides with short alkyl chains to a small extent. Esterases hydrolyzed the LEAD-alkylesters (3a-d) dependent on the chain length with τ = 55-82 min. Esters with branched alkyl chains were stable and introduction of the aromatic rings mentoined above increased the half-life to τ = 280 min and 360 min. In cell culture medium, only 3a-d degraded to 67-78 % after 72 h. However, the uptake studies showed that approximatly 80 % of the esters accumulated in the cell within the first 1-3 h of incubation. Therefore, it can be concluded that the intact LEAD-esters and LEAD-amides caused the biological effects. The compounds were non-cytotoxic and efficiently sensitized KD225 (K562-resistant) CML cells to Imatinib at a half-maximal sensitizing concentration (SC) of 1.5-2.9 μM (ester derivatives) and 1.3-11.2 μM (amide derivatives).
Diastereoselective synthesis and biological evaluation of new fluorine-containing α-aminophosphonates as anticancer agents and scaffold to human urokinase plasminogen activator inhibitors
Ciesielska K, Wawrzyniak D, Dutkiewicz G, Kubicki M, Jankowski W, Hoffmann M, Kamel K, Rolle K and Pluskota-Karwatka D
Phosphonate analogues of α-amino acids are increasingly valued for their significant potential in medicinal chemistry. Fluorine is a "magic" element that plays a huge role in modulating the properties of organic compounds. In this work, we combined the two pharmacophores in the synthesis of three series of new α-aminophosphonates. These compounds were obtained by diastereoselective hydrophosphonylation of imines prepared by an environmentally friendly mechanochemical approach. Results of computational SwissADME analysis suggested favorable drug-like properties of the α-aminophosphonates and indicated their potential for interaction with diverse biological targets including proteases, showing promising pharmacokinetic profiles compared to 5-fluoro-2'-deoxyuridine (FdU) used as a standard anticancer drug. Screening against ten cancer cell lines from seven types of cancer showed that five of the twenty compounds tested (1c, 2a, 2h, 3e, and 3f) exhibited superior activity against the HeLa cell line and lower cytotoxicity against normal MRC-5 cells than FdU. Compound 3e showed notable inhibitory effect on the MDA-MB-231 cell line, while 3a, 3h, and 3g demonstrated significant cytotoxic activity against U-87 MG and U-251 MG lines. Molecular docking highlighted the strong binding of compound 2a to the urokinase-type plasminogen activator (uPA) protein, with a binding affinity of -6.41 kcal/mol, suggesting the anti-metastatic potential of the compound. These findings enable to position the newly synthesized α-aminophosphonates as promising scaffolds for developing targeted anticancer therapies for metastatic cancers characterized by elevated uPA expression.
Photosensitizer formulations in photodynamic therapy of age-related macular degeneration
Beirão S, Pereira PMR, Fernandes R and Tomé JPC
Age-related macular degeneration (AMD) is a progressive degenerative disease that leads to visual impairment, predominantly affecting the elderly. Despite significant advancements in treatment, a definitive cure remains elusive. Current therapeutic strategies only slow down disease progression, inhibiting abnormal blood vessels growth, and preserving or improving vision. Among these strategies, photodynamic therapy (PDT) has emerged as a promising treatment, particularly for neovascular form, the most severe form of the disease. Although several photosensitizers (PS) have been developed, only one has received clinical approval for use in AMD. This treatment involves the intravenous administration of a photosensitizing agent that preferentially accumulates in the abnormal blood vessels beneath the macula. Upon activation by targeted laser light, the PS triggers photochemical reactions, leading to vascular occlusion and the reduction of choroidal neovascularization. This review provides a comprehensive overview of both experimental and clinical studies on PDT for AMD, discussing the current state of research, challenges in treatment optimization, and potential future directions to enhance this therapeutic approach.
Synthesis of 1-Hydroxy(and 1-Alkoxy, 1-Acyloxy)-1H-indoles and evaluations of their suppressive activities against tumor growth through inhibiting lactate dehydrogenase A
Cheon SY, Kim YE, Yang ES, Lim YJ, Bae CH, Jin JS, Park W, Kim BS, Kim C, Cho H, Kim S, Lee SH and Ha KT
Inhibition of lactate dehydrogenase (LDH) has emerged as a promising cancer therapy strategy due to its essential role in the metabolic transformation of cancer cells. In this study, 53 derivatives of 1-hydroxy(and 1-alkoxy, 1-acyloxy)indoles were designed, synthesized, and biologically evaluated. Several multi-substituted 1-hydroxy(and 1-alkoxy, 1-acyloxy)indole compounds exhibited inhibitory activity against the LDH-A isoform (LDHA). We confirmed that the C(3)-substituent provided additional significant hydrogen bonding and hydrophobic interactions, which enhanced the LDHA inhibitory activity with high selectivity. Our results revealed that methyl 4-bromo-3-[(n-hexyloxy)methyl]-1-hydroxy-1H-indole-2-carboxylate (1g), selectively inhibited LDHA (IC = 25 ± 1.12 nM) without affecting the LDH-B isoform (LDHB). The compound exhibited potent cytotoxic activity in several cancer cell lines, including DLD-1 colorectal cancer cells (GI = 27 ± 1.2 μM). Compound 1g significantly inhibited cancer cell growth by activating apoptotic pathways in a xenograft cancer model, without causing weight loss or liver and kidney damage. Therefore, compound 1g may serve as a highly specific and promising candidate for the development of LDHA inhibitors for cancer therapy.
Nanomolar activity of coumarin-3-thiosemicarbazones targeting Trypanosoma cruzi cruzain and the T. brucei cathepsin L-like protease
Nunes JA, Santos-Júnior PFDS, Gomes MC, Ferreira LAS, Padilha EKA, Teixeira TR, Stanger EJ, Kaur Y, Silva EBD, Costa CACB, Freitas JD, Araújo-Júnior JX, Mendonça-Junior FJB, Giardini MA, Siqueira-Neto JL, Caffrey CR, Zhan P, Cardoso SH and Silva-Júnior EFD
Trypanosoma cruzi (T. cruzi) and Trypanosoma brucei (T. brucei) urgently demand innovative drug development due to their impact on public health worldwide. Their cysteine proteases, Cruzain (CRZ) and the T. brucei Cathepsin L-like protease (TbrCATL) are established drug targets for these parasites. In this study, our coumarin-3-thiosemicarbazones demonstrated nanomolar IC values (22-698 nM) toward these proteases. Against T. cruzi amastigotes and T. brucei trypomastigotes, LASF-01 displayed a promising result. Herein, MCG-02, the most potent TbrCATL inhibitor, underwent comprehensive analyses, including cytotoxicity assessments and in vitro tests. Molecular dynamics (MD) simulations and a multiscale Quantum Mechanics/Quantum Mechanics (QM/QM) approach were used to generate insights into their binding modes. These suggested that MCG-02 could be a reversible, competitive covalent inhibitor. Further, confirmatory assays were experimentally performed changing different parameters to prove its efficacy. Additionally, the predicted pharmacokinetic profile showed that there is no violation of the Lipinski rule of five. Notably, coumarin-3-thiosemicarbazone hybrids emerged as promising candidates for designing highly active inhibitors against CRZ and TbrCATL. Overall, the integration of in silico and experimental approaches enhanced our understanding regarding the binding modes of MCG-02, which were experimentally corroborated, providing valuable insights for future drug development.