Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Acta Pharm Sin B ; 14(3): 1030-1076, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38487004

ABSTRACT

Synthetic chemistry plays an indispensable role in drug discovery, contributing to hit compounds identification, lead compounds optimization, candidate drugs preparation, and so on. As Nobel Prize laureate James Black emphasized, "the most fruitful basis for the discovery of a new drug is to start with an old drug"1. Late-stage modification or functionalization of drugs, natural products and bioactive compounds have garnered significant interest due to its ability to introduce diverse elements into bioactive compounds promptly. Such modifications alter the chemical space and physiochemical properties of these compounds, ultimately influencing their potency and druggability. To enrich a toolbox of chemical modification methods for drug discovery, this review focuses on the incorporation of halogen, oxygen, and nitrogen-the ubiquitous elements in pharmacophore components of the marketed drugs-through late-stage modification in recent two decades, and discusses the state and challenges faced in these fields. We also emphasize that increasing cooperation between chemists and pharmacists may be conducive to the rapid discovery of new activities of the functionalized molecules. Ultimately, we hope this review would serve as a valuable resource, facilitating the application of late-stage modification in the construction of novel molecules and inspiring innovative concepts for designing and building new drugs.

2.
Eur J Med Chem ; 269: 116323, 2024 Apr 05.
Article in English | MEDLINE | ID: mdl-38547735

ABSTRACT

Farnesoid X receptor (FXR) is a bile acids receptor and plays a crucial role in regulating bile acids, lipids, and glucose metabolism. Previous research suggests that inhibiting FXR activation can be beneficial in reducing cholesterol and low-density lipoprotein cholesterol (LDL-C) levels, offering potential treatment options for metabolic syndrome with lipid disorders. Herein, we report p-acetylaminobenzene sulfonate derivatives as a novel scaffold of FXR antagonists by multistage screening. Among these derivatives, compound F44-A13 exhibited a half-maximal inhibitory concentration of 1.1 µM. Furthermore, compound F44-A13 demonstrated effective inhibition of FXR activation in cellular assays and exhibited high selectivity over eleven other nuclear receptors. Besides, compound F44-A13 significantly suppressed the regulation of FXR target genes Shp, Besp, and Cyp7a1, while reducing cholesterol levels in human hepatoma HepG2 cells. Pharmacological studies conducted on C57BL/6 mice further confirmed that compound F44-A13 had beneficial effects in reducing cholesterol, triglycerides, and LDL-C levels. These findings highlight that F44-A13 is a highly selective FXR antagonist that might serve as a useful molecule for further FXR studies as well as the development of FXR antagonists for the potential treatment of metabolic diseases with lipid disorders.


Subject(s)
Bile Acids and Salts , Cholesterol , Mice , Animals , Humans , Cholesterol, LDL , Mice, Inbred C57BL , Structure-Activity Relationship , Cholesterol/metabolism , Bile Acids and Salts/pharmacology , Liver/metabolism
3.
Bioorg Med Chem Lett ; 97: 129547, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37944867

ABSTRACT

The COVID-19 caused by SARS-CoV-2 has led to a global pandemic that continues to impact societies and economies worldwide. The main protease (Mpro) plays a crucial role in SARS-CoV-2 replication and is an attractive target for anti-SARS-CoV-2 drug discovery. Herein, we report a series of 3-oxo-1,2,3,4-tetrahydropyrido[1,2-a]pyrazin derivatives as non-peptidomimetic inhibitors targeting SARS-CoV-2 Mpro through structure-based virtual screening and biological evaluation. Further similarity search and structure-activity relationship study led to the identification of compound M56-S2 with the enzymatic IC50 value of 4.0 µM. Moreover, the molecular simulation and predicted ADMET properties, indicated that non-peptidomimetic inhibitor M56-S2 might serve as a useful starting point for the further discovery of highly potent inhibitors targeting SARS-CoV-2 Mpro.


Subject(s)
COVID-19 , Pyrazines , SARS-CoV-2 , Humans , Antiviral Agents/pharmacology , COVID-19/prevention & control , Molecular Docking Simulation , Molecular Dynamics Simulation , Protease Inhibitors/pharmacology , SARS-CoV-2/drug effects , Viral Nonstructural Proteins , Pyrazines/chemistry , Pyrazines/pharmacology , COVID-19 Drug Treatment
4.
Nat Metab ; 5(10): 1726-1746, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37770763

ABSTRACT

Chronic inflammation due to islet-residing macrophages plays key roles in the development of type 2 diabetes mellitus. By systematically profiling intra-islet lipid-transmembrane receptor signalling in islet-resident macrophages, we identified endogenous 9(S)-hydroxy-10,12-octadecadienoic acid-G-protein-coupled receptor 132 (GPR132)-Gi signalling as a significant contributor to islet macrophage reprogramming and found that GPR132 deficiency in macrophages reversed metabolic disorders in mice fed a high-fat diet. The cryo-electron microscopy structures of GPR132 bound with two endogenous agonists, N-palmitoylglycine and 9(S)-hydroxy-10,12-octadecadienoic acid, enabled us to rationally design both GPR132 agonists and antagonists with high potency and selectivity through stepwise translational approaches. We ultimately identified a selective GPR132 antagonist, NOX-6-18, that modulates macrophage reprogramming within pancreatic islets, decreases weight gain and enhances glucose metabolism in mice fed a high-fat diet. Our study not only illustrates that intra-islet lipid signalling contributes to islet macrophage reprogramming but also provides a broadly applicable strategy for the identification of important G-protein-coupled receptor targets in pathophysiological processes, followed by the rational design of therapeutic leads for refractory diseases such as diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Islets of Langerhans , Mice , Animals , Diabetes Mellitus, Type 2/metabolism , Cryoelectron Microscopy , Islets of Langerhans/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
5.
Eur J Med Chem ; 250: 115167, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36764123

ABSTRACT

An indolin-2-(4-thiazolidinone) scaffold was previously shown to be a novel chemotype for JNK3 inhibition. However, more in vivo applications were limited due to the unconfirmed configuration and poor physicochemical properties. Here, the indolin-2-(4-thiazolidinone) scaffold validated the absolute configuration; substituents on the scaffold were optimized. Extensive structure activity relationship (SAR) studies were performed using kinase activity assays, thus leading to potent and highly selective JNK3 inhibitors with neuroprotective activity and good oral bioavailability. One lead compound, A53, was a potent and selective JNK3 inhibitor (IC50 = 78 nM) that had significant inhibition (>80% at 1 µM) to only JNK3 in a 398-kinase panel. A53 had low inhibition against JNK3 and high stability (t1/2(α) = 0.98 h, t1/2(ß) = 2.74 h) during oral administration. A modeling study of A53 in human JNK3 showed that the indolin-2-(4-thiazolidinone)-based JNK3 inhibitor with a 5-position-substituted hydrophilic group offered improved kinase inhibition.


Subject(s)
JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase 10 , Humans , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Isoforms
6.
J Am Chem Soc ; 144(29): 13415-13425, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35839515

ABSTRACT

The electrophilic halogenation of arenes is perhaps the simplest method to prepare aryl halides, which are important structural motifs in agrochemicals, materials, and pharmaceuticals. However, the nucleophilicity of arenes is weakened by the electron-withdrawing substituents, whose electrophilic halogenation reactions usually require harsh conditions and lead to limited substrate scopes and applications. Therefore, the halogenation of arenes containing electron-withdrawing groups (EWGs) and complex bioactive compounds under mild conditions has been a long-standing challenge. Herein, we describe Brønsted acid-catalyzed halogenation of arenes with electron-withdrawing substituents under mild conditions, providing an efficient protocol for aryl halides. The hydrogen bonding of Brønsted acid with the protic solvent 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP) enables this transformation and thus solves this long-standing problem.


Subject(s)
Electrons , Halogenation , Acids , Catalysis , Hydrogen Bonding
7.
Eur J Med Chem ; 238: 114508, 2022 Aug 05.
Article in English | MEDLINE | ID: mdl-35688005

ABSTRACT

The COVID-19 posed a serious threat to human life and health, and SARS-CoV-2 Mpro has been considered as an attractive drug target for the treatment of COVID-19. Herein, we report 2-(furan-2-ylmethylene)hydrazine-1-carbothioamide derivatives as novel inhibitors of SARS-CoV-2 Mpro developed by in-house library screening and biological evaluation. Similarity search led to the identification of compound F8-S43 with the enzymatic IC50 value of 10.76 µM. Further structure-based drug design and synthetic optimization uncovered compounds F8-B6 and F8-B22 as novel non-peptidomimetic inhibitors of Mpro with IC50 values of 1.57 µM and 1.55 µM, respectively. Moreover, enzymatic kinetic assay and mass spectrometry demonstrated that F8-B6 was a reversible covalent inhibitor of Mpro. Besides, F8-B6 showed low cytotoxicity with CC50 values of more than 100 µM in Vero and MDCK cells. Overall, these novel SARS-CoV-2 Mpro non-peptidomimetic inhibitors provide a useful starting point for further structural optimization.


Subject(s)
COVID-19 Drug Treatment , Coronavirus 3C Proteases , Furans , SARS-CoV-2 , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/chemistry , Coronavirus 3C Proteases/metabolism , Drug Discovery/methods , Furans/chemistry , Furans/pharmacology , Humans , Hydrazines/pharmacology , Molecular Docking Simulation , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology
8.
Eur J Med Chem ; 233: 114196, 2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35231830

ABSTRACT

Ataxia-telangiectasia mutated (ATM) kinase is a serine/threonine protein kinase and plays a key role in DNA double-strand breaks repair. Thus, ATM is considered a promising target for radiotherapy and chemotherapy sensitizing. Herein, we report the discovery of ATM agonist A22 and inhibitor A41 by computational methods and further biological evaluation. Among them, A22 exhibited low cytotoxicity in vitro and might serve as a useful tool for ATM research. Moreover, we firstly proved that ATM inhibitors could sensitize Irinotecan and Etoposide in a time-dependent manner on MCF-7 and SW480 cells, antagonism in a short period treatment while synergy at a long-term treatment and ATM agonist worked in an opposite way of ATM inhibitors. Further mechanism study demonstrated that the antagonism effect of ATM inhibitors with chemotherapeutic agents in a short period was resulting from inhibiting the p53/p21 axis to accelerate G1/S phase cell-cycle transition and promote cell survival. Additionally, A41 displayed antitumor effects combined with a chemotherapeutic drug in the SW480 xenograft model, indicating that A41 is a promising ATM inhibitor, which could increase the antitumor effect of chemotherapeutic drugs in vivo. All in all, these findings will guide the combination of ATM inhibitors with chemotherapeutic agents in further preclinical and clinical studies.


Subject(s)
Ataxia Telangiectasia , Neoplasms , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Humans , Phosphorylation , Protein Serine-Threonine Kinases
9.
Eur J Med Chem ; 201: 112445, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32603981

ABSTRACT

The c-Jun N-terminal kinase 3 (JNK3) plays key roles in a wide range of diseases, including neurodegeneration diseases, inflammation diseases, cancers, cardiovascular diseases, and metabolic disorders. Previously, we have identified a lead compound, (Z)-3-(2-(naphthalen-1-yl)-2-oxoethylidene)-3,4-dihydroquinoxalin-2(1H)-one (J46), which contains a 3,4-dihydroquinoxalin-2(1H)-one core structure as a key fragment to inhibit JNK3. However, compound J46 displayed high DDR1 and EGFR (T790M, L858R) inhibition and poor physicochemical properties, especially clogD and water-solubility, in its biological studies. Herein, we optimized compound J46 by structure-based drug design and exploiting the selectivity and physicochemical properties of various warhead groups to obtain compound J46-37, which not only exhibited a potent inhibition against JNK3 but also showed more than 50-fold potency better than DDR1 and EGFR (T790M, L858R). Furthermore, the selectivity and structure-activity relationship of novel synthesized 3,4-dihydroquinoxalin-2(1H)-one derivatives were analyzed by molecular docking and molecular dynamics simulation. Overall, compound J46-37, as a highly selective inhibitor of JNK3 with well physicochemical properties, is worth developing as therapies for the treatment of diseases related to JNK3.


Subject(s)
Mitogen-Activated Protein Kinase 10/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Quinoxalines/chemistry , Enzyme Assays , Humans , Mitogen-Activated Protein Kinase 10/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Quinoxalines/chemical synthesis , Quinoxalines/metabolism , Structure-Activity Relationship
10.
Org Biomol Chem ; 18(15): 2886-2892, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32236230

ABSTRACT

Terminal α-2,6-sialylation of N-glycans is a humanized glycosylation that affects the properties and efficacy of therapeutic glycoproteins. Fc di-sialylation (a biantennary N-glycan with two α-2,6-linked sialic acids) of IgG antibodies imparts them with enhanced anti-inflammatory activity and other roles. However, the microheterogeneity of N-glycoforms presents a challenge for therapeutic development. Therefore, controlled sialylation has drawn considerable attention, but direct access to well-defined di-sialylated antibodies remains limited. Herein, a one-pot three-enzyme protocol was developed by engineering a bacterial sialyltransferase to facilitate the modification of therapeutic antibodies with N-acetylneuraminic acid or its derivatives towards optimized glycosylation. To overcome the low proficiency of bacterial sialyltransferase in antibody remodeling, the Photobacterium sp. JT-ISH-224 α-2,6-sialyltransferase (Psp2,6ST) was genetically engineered by terminal truncation and site-directed mutagenesis based on its protein crystal structure. With the optimized reaction conditions and using activity-based screening of various Psp2,6ST variants, a truncated mutant Psp2,6ST (111-511)-His6 A235M/A366G was shown to effectively improve the catalytic efficiency of antibody di-sialylation. Herceptin and the donor substrate promiscuity allow the introduction of bioorthogonal modifications of N-acetylneuraminic acid into antibodies for site-specific conjugation. 2-AB hydrophilic interaction chromatography analysis of the released N-glycans and intact mass characterization confirmed the high di-sialylation of Herceptin via the optimized one-pot three-enzyme reaction. This study established a versatile enzymatic approach for producing highly di-sialylated IgG antibodies. It provides new insights into engineering bacterial sialyltransferase for adaptation to the enzymatic glycoengineering of therapeutic antibodies and the glycosite-specific conjugation of antibodies.


Subject(s)
Antibodies/metabolism , Photobacterium/enzymology , Protein Engineering , Sialic Acids/metabolism , Sialyltransferases/metabolism , Antibodies/chemistry , Sialyltransferases/genetics , beta-D-Galactoside alpha 2-6-Sialyltransferase
11.
Science ; 367(6475): 281-285, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31806697

ABSTRACT

The Schmidt reaction has been an efficient and widely used synthetic approach to amides and nitriles since its discovery in 1923. However, its application often entails the use of volatile, potentially explosive, and highly toxic azide reagents. Here, we report a sequence whereby triflic anhydride and formic and acetic acids activate the bulk chemical nitromethane to serve as a nitrogen donor in place of azides in Schmidt-like reactions. This protocol further expands the substrate scope to alkynes and simple alkyl benzenes for the preparation of amides and nitriles.

12.
J Med Chem ; 62(14): 6645-6664, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31268308

ABSTRACT

Alzheimer's disease (AD) is one of the most challenging diseases around the world with no effective clinical treatment. Previous studies have suggested c-Jun N-terminal kinase 3 (JNK3) as an attractive therapeutic target for AD. Herein, we report 3-substituted indolin-2-one derivatives as the first isoform-selective JNK3 inhibitors by multistage screening. In this study, comparative structure-based virtual screening was performed, and J30-8 was identified with a half-maximal inhibitory concentration of 40 nM, which exhibited over 2500-fold isoform selectivity and marked kinome-wide selectivity. Further study indicated that 1 µM J30-8 exhibited neuroprotective activity in vitro so as to alleviate the spatial memory impairment in vivo through reducing plaque burden and inhibiting the phosphorylation of JNKs, Aß precursor protein, and Tau protein. All of these indicated J30-8 as proved isoform-selective JNK3 inhibitors that might serve as a useful tool for further JNK3 studies with AD as well as for the development of JNK3 inhibitors for the potential treatment of neurodegenerative diseases.


Subject(s)
Indoles/chemistry , Indoles/pharmacology , Mitogen-Activated Protein Kinase 10/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Drug Design , Drug Discovery , Female , Humans , Indoles/pharmacokinetics , Mice , Mitogen-Activated Protein Kinase 10/metabolism , Models, Molecular , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacokinetics
13.
Eur J Med Chem ; 171: 221-234, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30925338

ABSTRACT

Glycogen synthase kinase 3α (GSK-3α) plays a constitutive role in various physiological processes and has been proved to be a therapeutic target for acute myeloid leukemia (AML). In this paper, by means of computer-aided drug design, we discovered a novel chemical series of GSK-3α inhibitors with an IC50 value of 0.033-2.804 µM. The preliminary structure-activity relationship was concluded and, notably, the most potent and isoform-selective compound G28_14 was identified with IC50 values of 33 nM and 218 nM against GSK-3α and -3ß, respectively, exhibiting a nearly ten-fold isoform-selectivity. Further cell viability assays and colony formation assays revealed that G28_14 suppressed cell survival by impairing cell proliferation by up to 90% in two AML cell lines. Moreover, surface marker expression analysis demonstrated that G28_14 induced terminal differentiation with a high level of CD11b, CD11c, and CD14. Western immunoblotting showed that G28_14 isoform-selectively inhibited the phosphorylation of GSK-3α in-cell without activating Wnt/ß-catenin signaling. In addition, to elucidate its structure-activity relationship, the binding mode of this chemical series was proposed using molecular docking and molecular dynamics simulations. Taken together, this chemical series is worth developing as differentiation therapies for the treatment of AML.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Glycogen Synthase Kinase 3/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Screening Assays, Antitumor , Glycogen Synthase Kinase 3/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Molecular Docking Simulation , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
14.
Eur J Med Chem ; 163: 583-596, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-30557831

ABSTRACT

Sulfated hyaluronic acid (sHA) is chemically synthetic mimetic of glycosaminoglycan (GAG) presenting promising biological functions. Specific sulfation pattern, termed as sulfation code plays critical roles in regulating the binding mode between GAG and proteins. As a structural analogue of chondroitin sulfate (CS), sHA bears much higher molecular weight and is nearly free of other proteoglycan contaminants. These properties make sHA a better bioscaffold to build safer and more functionalized material. However, chemical sulfonation process on naked HA polysaccharide produces random sulfation patterns which makes it difficult in disclosing the SAR. Herein, we utilized sHA and CS oligosaccharides with defined sulfation pattern to unravel the SAR between sHA and neurogenesis. We demonstrate sHA tetrasaccharide bearing 6-O-sulfation (sHA-6S) but not other sulfation patterns bind to growth factors at nanomolar range and promote the neurite outgrowth of rat E18 hippocampal neurons in vitro. Furthermore, synthetic sHA polysaccharide enriched in 6-O-sulfation also promote the hippocampal neurite outgrowth in vitro. Our work provides an effective method to disclose the bioactive sulfation pattern of sHA. Our results indicate that a specific sHA sulfation pattern could direct important physiological processes and open the way for the application of sHA-6S in neuroscience and medicine.


Subject(s)
Biomimetic Materials/chemistry , Glycosaminoglycans/chemistry , Hyaluronic Acid/pharmacology , Neurogenesis/drug effects , Sulfates/chemistry , Animals , Chondroitin Sulfates , Hippocampus/cytology , Hyaluronic Acid/chemistry , Neurons/ultrastructure , Oligosaccharides , Protein Binding , Rats
15.
Front Hum Neurosci ; 12: 417, 2018.
Article in English | MEDLINE | ID: mdl-30369874

ABSTRACT

Nowadays, the international community is becoming increasingly concerned about the sustainable utilization of natural resources. In order to protect the environment and reward sustainable practices, eco-labeling that signifies the environmental friendliness of the labeled food is already widely promoted in many regions around the world. Thus, it is of great importance for researchers to study consumers' attitudes toward eco-labeled food as food is supposed to satisfy consumers' needs. This study employed the event-related potentials (ERPs) approach to investigate consumers' attitudes toward eco-labeled food by comparing their neural processing of visual stimuli depicting eco-labeled and non-labeled food. Our results showed that behaviorally, participants preferred to buy eco-labeled food rather than non-labeled one. At the neural level, we observed markedly smaller P2 and N2 amplitudes when pictures of eco-labeled food were presented. Furthermore, we also found that amplitudes of P2 were negatively correlated with participants' purchase intention. Therefore, our current findings suggest that, while the environmental-friendly eco-labeling was not to one's own interests, it might still be evocative, which induce consumers' positive emotion, bring less cognitive conflict to the purchase decision-making and then result in a greater purchasing intention. This effect might be the result of the delivered value of social desirability.

16.
Bioorg Med Chem Lett ; 28(2): 160-166, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29208522

ABSTRACT

Glycogen synthase kinase-3ß (GSK-3ß) is an attractive therapeutic target for human diseases, such as diabetes, cancer, neurodegenerative diseases, and inflammation. Thus, structure-based virtual screening was performed to identify novel scaffolds of GSK-3ß inhibitors, and we observed that conserved water molecules of GSK-3ß were suitable for virtual screening. We found 14 hits and D1 (IC50 of 0.71 µM) were identified. Furthermore, the neuroprotection activity of D1-D3 was validated on a cellular level. 2D similarity searches were used to find derivatives of high inhibitory compounds and an enriched structure-activity relationship suggested that these skeletons were worthy of study as potent GSK-3ß inhibitors.


Subject(s)
Drug Discovery , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...