Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.140
1.
BMJ Open Ophthalmol ; 9(1)2024 May 03.
Article En | MEDLINE | ID: mdl-38702178

BACKGROUND: Dry eye disease is the most commonplace multifractional ocular complication, which has already affected millions of people in the world. It is identified by the excessive buildup of reactive oxygen species, leading to substantial corneal epithelial cell demise and ocular surface inflammation attributed to TLR4. In this study, we aimed to identify potential compounds to treat of dry eye syndrome by exploring in silico methods. METHODS: In this research, molecular docking and dynamics simulation tests were used to examine the effects of selected compounds on TLR4 receptor. Compounds were extracted from different databases and were prepared and docked against TLR4 receptor via Autodock Vina. Celastrol, lumacaftor and nilotinib were selected for further molecular dynamics studies for a deeper understanding of molecular systems consisting of protein and ligands by using the Desmond module of the Schrodinger Suite. RESULTS: The docking results revealed that the compounds are having binding affinity in the range of -5.1 to -8.78 based on the binding affinity and three-dimensional interactions celastrol, lumacaftor and nilotinib were further studied for their activity by molecular dynamics. Among the three compounds, celastrol was the most stable based on molecular dynamics trajectory analysis from 100 ns in the catalytic pockets of 2Z63.pdb.pdb. Root mean square deviation of celastrol/2Z63 was in the range of 1.8-4.8 Å. CONCLUSION: In particular, Glu376 of TLR4 receptor is crucial for the identification and binding of lipopolysaccharides (LPS), which are part of Gram-negative bacteria's outer membrane. In our investigation, celastrol binds to Glu376, suggesting that celastrol may prevent the dry eye syndrome by inhibiting LPS's binding to TLR4.


Dry Eye Syndromes , Molecular Docking Simulation , Molecular Dynamics Simulation , Pentacyclic Triterpenes , Pyrimidines , Toll-Like Receptor 4 , Dry Eye Syndromes/drug therapy , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/chemistry , Humans , Pentacyclic Triterpenes/pharmacology , Pentacyclic Triterpenes/chemistry , Pentacyclic Triterpenes/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/therapeutic use , Triterpenes/pharmacology , Triterpenes/chemistry , Computer Simulation , Ligands , Aminopyridines/pharmacology , Aminopyridines/chemistry , Aminopyridines/therapeutic use
2.
Chem Biol Drug Des ; 103(5): e14530, 2024 May.
Article En | MEDLINE | ID: mdl-38725091

Feline immunodeficiency virus (FIV) is a common infection found in domesticated and wild cats worldwide. Despite the wealth of therapeutic understanding of the disease in humans, considerably less information exists regarding the treatment of the disease in felines. Current treatment relies on drugs developed for the related human immunodeficiency virus (HIV) and includes compounds of the popular non-nucleotide reverse transcriptase (NNRTI) class. This is despite FIV-RT being only 67% similar to HIV-1 RT at the enzyme level, increasing to 88% for the allosteric pocket targeted by NNRTIs. The goal of this project was to try to quantify how well the more extensive pharmacological knowledge available for human disease translates to felines. To this end we screened known NNRTIs and 10 diverse pyrimidine analogs identified virtually. We use this chemo-centric probe approach to (a) assess the similarity between the two related RT targets based on the observed experimental inhibition values, (b) try to identify more potent inhibitors at FIV, and (c) gain a better appreciation of the structure-activity relationships (SAR). We found the correlation between IC50s at the two targets to be strong (r2 = 0.87) and identified compound 1 as the most potent inhibitor of FIV with IC50 of 0.030 µM ± 0.009. This compared to FIV IC50 values of 0.22 ± 0.17 µM, 0.040 ± 0.010 µM and >160 µM for known anti HIV-1 RT drugs Efavirenz, Rilpivirine, and Nevirapine, respectively. This knowledge, along with an understanding of the structural origin that give rise to any differences could improve the way HIV drugs are repurposed for FIV.


HIV Reverse Transcriptase , Immunodeficiency Virus, Feline , Reverse Transcriptase Inhibitors , Animals , Reverse Transcriptase Inhibitors/pharmacology , Reverse Transcriptase Inhibitors/chemistry , Cats , Immunodeficiency Virus, Feline/drug effects , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/metabolism , Humans , Structure-Activity Relationship , Pyrimidines/chemistry , Pyrimidines/pharmacology , Alkynes/chemistry , Alkynes/pharmacology , HIV-1/drug effects , HIV-1/enzymology , Cyclopropanes/pharmacology , Cyclopropanes/chemistry , Molecular Docking Simulation , Benzoxazines/chemistry , Benzoxazines/pharmacology
3.
Pak J Pharm Sci ; 37(1(Special)): 173-184, 2024 Jan.
Article En | MEDLINE | ID: mdl-38747267

Hydrazones 1-6, azo-pyrazoles 7-9 and azo-pyrimidines 10-15 are compounds that exhibit antibacterial activity. The mode of action and structures of these derivatives have been previously confirmed as antibacterial. In this investigation, biological screening and molecular docking studies were performed for derivatives 1-15, with compounds 2, 7, 8, 14 and 15 yielding the best energy scores (from -20.7986 to -10.5302 kcal/mol). Drug-likeness and in silico ADME prediction for the most potent derivatives, 2, 7, 8, 14 and 15, were predicted (from 84.46 to 96.85%). The latter compounds showed good recorded physicochemical properties and pharmacokinetics. Compound 8 demonstrated the strongest inhibition, which was similar to the positive control (eflornithine) against Trypanosoma brucei brucei (WT), with an EC50 of 25.12 and 22.52µM, respectively. Moreover, compound 14 exhibited the best activity against Leishmania mexicana promastigotes and Leishmania major promastigotes (EC50 =46.85; 40.78µM, respectively).


Molecular Docking Simulation , Pyrazoles , Pyrimidines , Trypanocidal Agents , Trypanosoma brucei brucei , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Trypanosoma brucei brucei/drug effects , Pyrazoles/pharmacology , Pyrazoles/chemistry , Trypanocidal Agents/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/chemical synthesis , Leishmania mexicana/drug effects , Leishmania major/drug effects , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/chemistry , Computer Simulation , Azo Compounds/pharmacology , Azo Compounds/chemistry , Azo Compounds/chemical synthesis , Structure-Activity Relationship , Parasitic Sensitivity Tests
4.
J Am Chem Soc ; 146(19): 13317-13325, 2024 May 15.
Article En | MEDLINE | ID: mdl-38700457

We describe the synthesis and biological testing of ruthenium-bipyridine ruxolitinib (RuBiRuxo), a photoreleasable form of ruxolitinib, a JAK inhibitor used as an antitumoral agent in cutaneous T-cell lymphomas (CTCL). This novel caged compound is synthesized efficiently, is stable in aqueous solution at room temperature, and is photoreleased rapidly by visible light. Irradiation of RuBiRuxo reduces cell proliferation and induces apoptosis in a light- and time-dependent manner in a CTCL cell line. This effect is specific and is mediated by a decreased phosphorylation of STAT proteins. Our results demonstrate the potential of ruthenium-based photocompounds and light-based therapeutic approaches for the potential treatment of cutaneous lymphomas and other pathologies.


Antineoplastic Agents , Apoptosis , Cell Proliferation , Nitriles , Pyrazoles , Pyrimidines , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Nitriles/chemistry , Nitriles/pharmacology , Nitriles/chemical synthesis , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/chemical synthesis , Apoptosis/drug effects , Pyrazoles/pharmacology , Pyrazoles/chemistry , Pyrazoles/chemical synthesis , Cell Line, Tumor , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/chemical synthesis , Ruthenium/chemistry , Ruthenium/pharmacology , Light , Molecular Structure , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism
5.
AAPS PharmSciTech ; 25(5): 97, 2024 May 06.
Article En | MEDLINE | ID: mdl-38710894

Pancreatic ductal adenocarcinoma (PDAC) is one of the highly fatal types of cancer with high mortality/incidence. Considering the crucial role of vascular endothelial growth factor (VEGF) in PDAC progression, its inhibition can be a viable strategy for the treatment. Pazopanib, a second-generation VEGF inhibitor, is approved for the treatment of various oncological conditions. However, due to associated limitations like low oral bioavailability (14-39%), high inter/intra-subject variability, stability issues, etc., high doses (800 mg) are required, which further lead to non-specific toxicities and also contribute toward cancer resistance. Thus, to overcome these challenges, pazopanib-loaded PEGylated nanoliposomes were developed and evaluated against pancreatic cancer cell lines. The nanoliposomes were prepared by thin-film hydration method, followed by characterization and stability studies. This QbD-enabled process design successfully led to the development of a suitable pazopanib liposomal formulation with desirable properties. The % entrapment of PZP-loaded non-PEGylated and PEGylated nanoliposomes was found to be 75.2% and 84.9%, respectively, whereas their particle size was found to be 129.7 nm and 182.0 nm, respectively. The developed liposomal formulations exhibited a prolonged release and showed desirable physicochemical properties. Furthermore, these liposomal formulations were also assessed for in vitro cell lines, such as cell cytotoxicity assay and cell uptake. These studies confirm the effectiveness of developed liposomal formulations against pancreatic cancer cell lines. The outcomes of this work provide encouraging results and a way forward to thoroughly investigate its potential for PDAC treatment.


Carcinoma, Pancreatic Ductal , Indazoles , Liposomes , Nanoparticles , Pancreatic Neoplasms , Particle Size , Pyrimidines , Sulfonamides , Indazoles/administration & dosage , Indazoles/pharmacology , Humans , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Sulfonamides/chemistry , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Cell Line, Tumor , Pancreatic Neoplasms/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Drug Liberation , Chemistry, Pharmaceutical/methods
6.
Molecules ; 29(9)2024 May 03.
Article En | MEDLINE | ID: mdl-38731613

Ribonuclease H (RNase H) was identified as an important target for HIV therapy. Currently, no RNase H inhibitors have reached clinical status. Herein, a series of novel thiazolone[3,2-a]pyrimidine-containing RNase H inhibitors were developed, based on the hit compound 10i, identified from screening our in-house compound library. Some of these derivatives exhibited low micromolar inhibitory activity. Among them, compound 12b was identified as the most potent inhibitor of RNase H (IC50 = 2.98 µM). The experiment of magnesium ion coordination was performed to verify that this ligand could coordinate with magnesium ions, indicating its binding ability to the catalytic site of RNase H. Docking studies revealed the main interactions of this ligand with RNase H. A quantitative structure activity relationship (QSAR) was also conducted to disclose several predictive mathematic models. A molecular dynamics simulation was also conducted to determine the stability of the complex. Taken together, thiazolone[3,2-a]pyrimidine can be regarded as a potential scaffold for the further development of RNase H inhibitors.


Anti-HIV Agents , Molecular Docking Simulation , Pyrimidines , Quantitative Structure-Activity Relationship , Pyrimidines/chemistry , Pyrimidines/pharmacology , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Anti-HIV Agents/chemical synthesis , Humans , Molecular Dynamics Simulation , Ribonuclease H/antagonists & inhibitors , Ribonuclease H/metabolism , Drug Design , HIV Infections/drug therapy , HIV-1/drug effects , HIV-1/enzymology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Thiazoles/chemistry , Thiazoles/pharmacology , Molecular Structure
7.
Eur J Med Chem ; 271: 116395, 2024 May 05.
Article En | MEDLINE | ID: mdl-38626523

The transforming growth factor ß1 (TGFß1)/SMAD signaling pathway regulates many vital physiological processes. The development of potent inhibitors targeting activin receptor-like kinase 5 (ALK5) would provide potential treatment reagents for various diseases. A significant number of ALK5 inhibitors have been discovered, and they are currently undergoing clinical evaluation at various stages. However, the clinical demands were far from being met. In this study, we utilized an alternative conformation-similarity-based virtual screening (CSVS) combined with a fragment-based drug designing (FBDD) strategy to efficiently discover a potent and active hit with a novel chemical scaffold. After structural optimization in the principle of group replacement, compound 57 was identified as the most promising ALK5 inhibitor. Compound 57 demonstrated significant inhibitory effects against the TGF-ß1/SMAD signaling pathway. It could markedly attenuate the production of extracellular matrix (ECM) and deposition of collagen. Also, the lead compound showed adequate pharmacokinetic (PK) properties and good in vivo tolerance. Moreover, treatment with compound 57 in two different xerograph models showed significant inhibitory effects on the growth of pancreatic cancer cells. These results suggested that lead compound 57 refers as a promising ALK5 inhibitor both in vitro and in vivo, which merits further validation.


Drug Design , Protein Kinase Inhibitors , Pyrazoles , Pyrimidines , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/metabolism , Humans , Pyrazoles/pharmacology , Pyrazoles/chemistry , Pyrazoles/chemical synthesis , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Structure-Activity Relationship , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Animals , Molecular Structure , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Mice , Cell Line, Tumor , Drug Screening Assays, Antitumor , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism
8.
J Med Chem ; 67(9): 7245-7259, 2024 May 09.
Article En | MEDLINE | ID: mdl-38635563

Cofactor mimicry represents an attractive strategy for the development of enzyme inhibitors but can lead to off-target effects due to the evolutionary conservation of binding sites across the proteome. Here, we uncover the ADP-ribose (ADPr) hydrolase NUDT5 as an unexpected, noncovalent, off-target of clinical BTK inhibitors. Using a combination of biochemical, biophysical, and intact cell NanoBRET assays as well as X-ray crystallography, we confirm catalytic inhibition and cellular target engagement of NUDT5 and reveal an unusual binding mode that is independent of the reactive acrylamide warhead. Further investigation of the prototypical BTK inhibitor ibrutinib also revealed potent inhibition of the largely unstudied NUDIX hydrolase family member NUDT14. By exploring structure-activity relationships (SARs) around the core scaffold, we identify a potent, noncovalent, and cell-active dual NUDT5/14 inhibitor. Cocrystallization experiments yielded new insights into the NUDT14 hydrolase active site architecture and inhibitor binding, thus providing a basis for future chemical probe design.


Agammaglobulinaemia Tyrosine Kinase , Pyrophosphatases , Humans , Pyrophosphatases/antagonists & inhibitors , Pyrophosphatases/metabolism , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/metabolism , Structure-Activity Relationship , Crystallography, X-Ray , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/chemical synthesis , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrazoles/chemical synthesis , Pyrazoles/metabolism , Piperidines/pharmacology , Piperidines/chemistry , Piperidines/metabolism , Piperidines/chemical synthesis , Drug Discovery , Pyrimidines/chemistry , Pyrimidines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/metabolism , Adenine/analogs & derivatives , Adenine/chemistry , Adenine/pharmacology , Adenine/metabolism , Models, Molecular , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/chemical synthesis
9.
Eur J Med Chem ; 271: 116399, 2024 May 05.
Article En | MEDLINE | ID: mdl-38640868

The structural optimization of B14, an antibacterial agent we previously obtained, has led to the discovery of a new class of CH2-linked quinolone-aminopyrimidine hybrids with potent anti-MRSA activities. Surprisingly, the hybrids lacking a C-6 fluoro atom at the quinolone nucleus showed equal or even stronger anti-MRSA activities than their corresponding 6-fluoro counterparts, despite the well-established structure-activity relationships (SARs) indicating that the 6-fluoro substituent enhances the antibacterial activity in conventional fluoroquinolone antibiotics. Moreover, these new hybrids, albeit structurally related to conventional fluoroquinolones, showed no cross-resistance with fluoroquinolone drugs. The most active compound, 15m, exhibited excellent activities with a MIC value of 0.39 µg/mL against both fluoroquinolone-sensitive strain USA500 and -resistant MRSA isolate Mu50. Further resistance development studies indicated MRSA is unlikely to acquire resistance against 15m. Moreover, 15m displayed favorable in vivo half-life and safety profiles. These findings suggest a rationale for further evolution of quinolone antibiotics with a high barrier to resistance.


Anti-Bacterial Agents , Fluoroquinolones , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Pyrimidines , Quinolones , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Methicillin-Resistant Staphylococcus aureus/drug effects , Structure-Activity Relationship , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Fluoroquinolones/pharmacology , Fluoroquinolones/chemistry , Fluoroquinolones/chemical synthesis , Quinolones/pharmacology , Quinolones/chemistry , Quinolones/chemical synthesis , Molecular Structure , Drug Resistance, Bacterial/drug effects , Dose-Response Relationship, Drug , Animals , Humans
10.
Eur J Med Chem ; 271: 116391, 2024 May 05.
Article En | MEDLINE | ID: mdl-38669909

LIM Kinases, LIMK1 and LIMK2, have become promising targets for the development of inhibitors with potential application for the treatment of several major diseases. LIMKs play crucial roles in cytoskeleton remodeling as downstream effectors of small G proteins of the Rho-GTPase family, and as major regulators of cofilin, an actin depolymerizing factor. In this article we describe the conception, synthesis, and biological evaluation of novel tetrahydropyridine pyrrolopyrimidine LIMK inhibitors. Homology models were first constructed to better understand the binding mode of our preliminary compounds and to explain differences in biological activity. A library of over 60 products was generated and in vitro enzymatic activities were measured in the mid to low nanomolar range. The most promising derivatives were then evaluated in cell on cofilin phosphorylation inhibition which led to the identification of 52 which showed excellent selectivity for LIMKs in a kinase selectivity panel. We also demonstrated that 52 affected the cell cytoskeleton by disturbing actin filaments. Cell migration studies with this derivative using three different cell lines displayed a significant effect on cell motility. Finally, the crystal structure of the kinase domain of LIMK2 complexed with 52 was solved, greatly improving our understanding of the interaction between 52 and LIMK2 active site. The reported data represent a basis for the development of more efficient LIMK inhibitors for future in vivo preclinical validation.


Lim Kinases , Protein Kinase Inhibitors , Lim Kinases/antagonists & inhibitors , Lim Kinases/metabolism , Humans , Structure-Activity Relationship , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Molecular Structure , Cell Movement/drug effects , Models, Molecular , Pyridines/pharmacology , Pyridines/chemistry , Pyridines/chemical synthesis , Dose-Response Relationship, Drug , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/chemical synthesis
11.
J Med Chem ; 67(8): 6425-6455, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38613499

The RAS-RAF-MEK-ERK signaling cascade is abnormally activated in various tumors, playing a crucial role in mediating tumor progression. As the key component at the terminal stage of this cascade, ERK1/2 emerges as a potential antitumor target and offers a promising therapeutic strategy for tumors harboring BRAF or RAS mutations. Here, we identified 36c with a (thiophen-3-yl)aminopyrimidine scaffold as a potent ERK1/2 inhibitor through structure-guided optimization for hit 18. In preclinical studies, 36c showed powerful ERK1/2 inhibitory activities (ERK1/2 IC50 = 0.11/0.08 nM) and potent antitumor efficacy both in vitro and in vivo against triple-negative breast cancer and colorectal cancer models harboring BRAF and RAS mutations. 36c could directly inhibit ERK1/2, significantly block the phosphorylation expression of their downstream substrates p90RSK and c-Myc, and induce cell apoptosis and incomplete autophagy-related cell death. Taken together, this work provides a promising ERK1/2 lead compound for multiple tumor-treatment drug discovery.


Antineoplastic Agents , Protein Kinase Inhibitors , Pyrimidines , Humans , Pyrimidines/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Structure-Activity Relationship , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Thiophenes/pharmacology , Thiophenes/chemical synthesis , Thiophenes/chemistry , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Cell Line, Tumor , Drug Discovery , Apoptosis/drug effects , Female , Mice, Nude , Drug Screening Assays, Antitumor , Molecular Structure , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Mice, Inbred BALB C
12.
Int J Pharm ; 656: 124108, 2024 May 10.
Article En | MEDLINE | ID: mdl-38604540

Lipid-based formulations (LbFs) are an extensively used approach for oral delivery of poorly soluble drug compounds in the form of lipid suspension and lipid solution. However, the high target dose and inadequate lipid solubility limit the potential of brick dust molecules to be formulated as LbFs. Thus, the complexation of such molecules with a lipophilic counterion can be a plausible approach to improve the solubility in lipid-based solutions via reducing drug crystallinity and polar surface area. The study aimed to enhance drug loading in lipid solution for Nilotinib (Nil) through complexation or salt formation with different lipophilic counterions. We synthesized different lipophilic salts/ complexes via metathesis reactions and confirmed their formation by 1H NMR and FTIR. Docusate-based lipophilic salt showed improved solubility in medium-chain triglycerides (∼7 to 7.5-fold) and long-chain triglycerides (∼30 to 35-fold) based lipids compared to unformulated crystalline Nil. The increased lipid solubility could be attributed to the reduction in drug crystallinity which was further confirmed by the PXRD and DSC. Prototype LbFs were prepared to evaluate drug loading and their physicochemical characteristics. The findings suggested that structural features of counterion including chain length and lipophilicity affect the drug loading in LbF. In addition, physical stability testing of formulations was performed, inferring that aliphatic sulfate-based LbFs were stable with no sign of drug precipitation or salt disproportionation. An in vitro lipolysis-permeation study revealed that the primary driver of absorptive flux is the solubilization of the drug and reduced amount of lipid. Further, the in vivo characterization was conducted to measure the influence of increased drug load on oral bioavailability. Overall, the results revealed enhanced absorption of lipophilic salt-based LbF over unformulated crystalline Nil and conventional LbF (drug load equivalent to equilibrium solubility) which supports the idea that lipophilic salt-based LbF enhances drug loading, and supersaturation-mediated drug solubilization, unlocking the full potential of LbF.


Lipids , Salts , Solubility , Salts/chemistry , Animals , Lipids/chemistry , Male , Administration, Oral , Drug Compounding/methods , Pyrimidines/chemistry , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Dust , Drug Liberation , Rats , Chemistry, Pharmaceutical/methods , Hydrophobic and Hydrophilic Interactions , Triglycerides/chemistry , Drug Stability , Drug Carriers/chemistry , Crystallization
13.
J Med Chem ; 67(8): 6052-6063, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38592948

Akt kinase is vital in cell growth, survival, metabolism, and migration. Dysregulation of Akt signaling is implicated in cancer and metabolic disorders. In the context of cancer, overactive Akt promotes cell survival and proliferation. This has spurred extensive research into developing Akt inhibitors as potential therapeutic agents to disrupt aberrant Akt signaling. Akt inhibitors are classified into three main types: ATP-competitive, allosteric, and covalent-allosteric inhibitors (CAAIs). ATP-competitive inhibitors compete with ATP for binding to Akt, allosteric inhibitors interact with the Pleckstrin homology (PH) domain, and covalent-allosteric inhibitors form covalent bonds, making them more potent and selective. Notably, capivasertib (AZD5363), a potent ATP-competitive Akt inhibitor, received FDA approval in November 2023 for use in combination with the estrogen receptor degrader fulvestrant to treat breast cancer. Challenges remain, including improving selectivity, identifying biomarkers to tailor treatments, and enhancing therapeutic efficacy while minimizing adverse effects. Particularly covalent-allosteric inhibitors hold promise for future more effective and personalized treatments.


Protein Kinase Inhibitors , Proto-Oncogene Proteins c-akt , Pyrimidines , Humans , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/therapeutic use , Allosteric Regulation/drug effects , Drug Approval , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Pyrroles/chemistry , Pyrroles/pharmacology , Pyrroles/chemical synthesis , Animals
14.
Org Biomol Chem ; 22(16): 3249-3261, 2024 04 24.
Article En | MEDLINE | ID: mdl-38568016

A one-pot microwave assisted telescopic approach is reported for the chemo-selective synthesis of substituted 1,3-thiazetidines using readily available 2-aminopyridines/pyrazines/pyrimidine, substituted isothiocyanates and 1,2-dihalomethanes. The procedure involves thiourea formation from 2-aminopyridines/pyrazines/pyrimidine with the substituted isothiocyanates followed by a base catalysed nucleophilic attack of the CS bond on the 1,2-dihalomethane. Subsequently, a cyclization reaction occurs to yield substituted 1,3-thiazetidines. These four membered strained ring systems are reported to possess broad substrate scope with high functional group tolerance. The above synthetic sequence for the formation of four membered heterocycles is proven to be a modular and straightforward approach. Further the mechanistic pathway for the formation of 1,3-thiazetidines was supported by computational evaluations and X-ray crystallography analyses. The relevance of these thiazetidines in biological applications is evaluated by studying their ability to bind bio-macromolecules like proteins and nucleic acids.


Microwaves , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Crystallography, X-Ray , Proteins/chemistry , Thiazoles/chemistry , Thiazoles/chemical synthesis , Models, Molecular , Molecular Structure , Nucleic Acids/chemistry , Nucleic Acids/chemical synthesis , Isothiocyanates/chemistry , Isothiocyanates/chemical synthesis , Aminopyridines/chemistry , Aminopyridines/chemical synthesis
15.
Luminescence ; 39(5): e4744, 2024 May.
Article En | MEDLINE | ID: mdl-38682162

Hydrazine substituted thienopyrimidine, a new fluorophore, was used to synthesize a novel Schiff base R1 as a chemosensor via the condensation with p-formyltriphenylamine, and the structure was confirmed using nuclear magnetic resonance spectroscopy (NMR) and mass spectrometry (MS) analysis. When treated with Cu2+ in dimethylsulfoxide (DMSO)/H2O buffer, R1 showed a phenomenon of fluorescence quenching, which was reversible with the action of ethylenediaminetetraacetic acid (EDTA). When treated with Fe3+ in dimethylformamide (DMF)/H2O buffer, R1 exhibited the same phenomenon, but fluorescence was recovered with inorganic pyrophosphate (PPi) quantitatively. The complexation ratios for R1-Cu2+ and R1-Fe3+ were both 1:2, which were manifested by MS titrations and corresponding Job's plots. The limits of detection of R1 for Cu2+ and Fe3+ were 3.11 × 10-8 and 1.24 × 10-7 M, respectively. The sensing mechanism of R1 toward Cu2+ and Fe3+ was confirmed using density functional theory calculations and electrostatic potential analysis. Test strips of R1 were fabricated successfully for on-site detection of Cu2+ and Fe3+. In addition, R1 was applied to recognize Cu2+ and Fe3+ in actual water samples with satisfactory recovery.


Copper , Diphosphates , Fluorescent Dyes , Iron , Pyrimidines , Solvents , Spectrometry, Fluorescence , Copper/chemistry , Copper/analysis , Pyrimidines/chemistry , Pyrimidines/analysis , Diphosphates/analysis , Diphosphates/chemistry , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Iron/analysis , Iron/chemistry , Solvents/chemistry , Molecular Structure , Fluorescence , Density Functional Theory
16.
Drug Dev Res ; 85(3): e22193, 2024 May.
Article En | MEDLINE | ID: mdl-38685605

The scaffolds of two known CDK inhibitors (CAN508 and dinaciclib) were the starting point for synthesizing two series of pyarazolo[1,5-a]pyrimidines to obtain potent inhibitors with proper selectivity. The study presented four promising compounds; 10d, 10e, 16a, and 16c based on cytotoxic studies. Compound 16a revealed superior activity in the preliminary anticancer screening with GI % = 79.02-99.13 against 15 cancer cell lines at 10 µM from NCI full panel 60 cancer cell lines and was then selected for further investigation. Furthermore, the four compounds revealed good safety profile toward the normal cell lines WI-38. These four compounds were subjected to CDK inhibitory activity against four different isoforms. All of them showed potent inhibition against CDK5/P25 and CDK9/CYCLINT. Compound 10d revealed the best activity against CDK5/P25 (IC50 = 0.063 µM) with proper selectivity index against CDK1 and CDK2. Compound 16c exhibited the highest inhibitory activity against CDK9/CYCLINT (IC50 = 0.074 µM) with good selectivity index against other isoforms. Finally, docking simulations were performed for compounds 10e and 16c accompanied by molecular dynamic simulations to understand their behavior in the active site of the two CDKs with respect to both CAN508 and dinaciclib.


Antineoplastic Agents , Bridged Bicyclo Compounds, Heterocyclic , Cyclic N-Oxides , Drug Design , Indolizines , Molecular Docking Simulation , Protein Kinase Inhibitors , Pyridinium Compounds , Humans , Pyridinium Compounds/pharmacology , Pyridinium Compounds/chemistry , Indolizines/pharmacology , Indolizines/chemistry , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Cyclic N-Oxides/pharmacology , Cyclic N-Oxides/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cyclin-Dependent Kinases/antagonists & inhibitors , Structure-Activity Relationship , Pyrimidines/pharmacology , Pyrimidines/chemistry , Drug Screening Assays, Antitumor , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/metabolism , Cyclin-Dependent Kinase 9/antagonists & inhibitors , Cyclin-Dependent Kinase 9/metabolism
17.
Org Biomol Chem ; 22(17): 3510-3517, 2024 05 01.
Article En | MEDLINE | ID: mdl-38619422

Post-synthetic conversion of the trifluoromethyl group to a heteroaryl group at the C5 position of the pyrimidine base in DNA oligonucleotides was achieved. Specifically, the oligonucleotides containing 5-trifluoromethylpyrimidine bases were treated with o-phenylenediamines and o-aminothiophenols as nucleophiles to afford the corresponding 5-(benzimidazol-2-yl)- and 5-(benzothiazol-2-yl)-pyrimidine-modified bases. Furthermore, evaluation of the fluorescence properties of the obtained oligonucleotides revealed that among them the oligonucleotide containing 5-(5-methylbenzimidazol-2-yl)cytosine exhibited the highest fluorescence intensity. These results indicated that post-synthetic trifluoromethyl conversion, which is practical and operationally simple, is a powerful tool for exploring functional oligonucleotides.


Fluorescent Dyes , Oligonucleotides , Pyrimidines , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Oligonucleotides/chemistry , Oligonucleotides/chemical synthesis , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , Molecular Structure
18.
Phys Chem Chem Phys ; 26(17): 13420-13431, 2024 May 01.
Article En | MEDLINE | ID: mdl-38647171

Autoimmune inflammatory diseases, such as rheumatoid arthritis (RA) and ulcerative colitis, are associated with an uncontrolled production of cytokines leading to the pronounced inflammatory response of these disorders. Their therapy is currently focused on the inhibition of cytokine receptors, such as the Janus kinase (JAK) protein family. Tofacitinib and peficitinib are JAK inhibitors that have been recently approved to treat rheumatoid arthritis. In this study, an in-depth analysis was carried out through quantum biochemistry to understand the interactions involved in the complexes formed by JAK1 and tofacitinib or peficitinib. Computational analyses provided new insights into the binding mechanisms between tofacitinib or peficitinib and JAK1. The essential amino acid residues that support the complex are also identified and reported. Additionally, we report new interactions, such as van der Waals; hydrogen bonds; and alkyl, pi-alkyl, and pi-sulfur forces, that stabilize the complexes. The computational results revealed that peficitinib presents a similar affinity to JAK1 compared to tofacitinib based on their interaction energies.


Adamantane/analogs & derivatives , Janus Kinase 1 , Niacinamide , Niacinamide/analogs & derivatives , Piperidines , Pyrimidines , Pyrimidines/chemistry , Pyrimidines/pharmacology , Piperidines/chemistry , Piperidines/pharmacology , Piperidines/therapeutic use , Niacinamide/chemistry , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/metabolism , Janus Kinase 1/chemistry , Humans , Quantum Theory , Autoimmune Diseases/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Hydrogen Bonding , Janus Kinase Inhibitors/chemistry , Janus Kinase Inhibitors/therapeutic use , Janus Kinase Inhibitors/pharmacology , Adamantane/chemistry , Pyrroles/chemistry , Pyrroles/pharmacology , Molecular Docking Simulation
19.
J Med Chem ; 67(9): 7620-7634, 2024 May 09.
Article En | MEDLINE | ID: mdl-38634707

Meisoindigo (Mei) has long been recognized in chronic myeloid leukemia (CML) treatment. To elucidate its molecular target and mechanisms, we embarked on designing and synthesizing a series of Mei-derived PROTACs. Through this endeavor, VHL-type PROTAC 9b was identified to be highly cytotoxic against SW620, SW480, and K562 cells. Employing DiaPASEF-based quantitative proteomic analysis, in combination with extensive validation assays, we unveiled that 9b potently and selectively degraded ATM across SW620 and SW480 cells in a ubiquitin-proteasome-dependent manner. 9b-induced selective ATM degradation prompted DNA damage response cascades, thereby leading to the cell cycle arrest and cell apoptosis. This pioneering discovery renders the advent of ATM degradation for anti-cancer therapy. Notably, 9b-induced ATM degradation synergistically enhanced the efficacy of ATR inhibitor AZD6738 both in vitro and in vivo. This work establishes the synthetic lethality-inducing properties of ATR inhibitors in the ATM-deficient context, thereby providing new avenues to innovative therapies for colorectal cancer.


Antineoplastic Agents , Ataxia Telangiectasia Mutated Proteins , Colorectal Neoplasms , Animals , Humans , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Drug Discovery , Indoles/pharmacology , Indoles/chemistry , Indoles/chemical synthesis , Mice, Nude , Proteolysis/drug effects , Pyrimidines/pharmacology , Pyrimidines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/therapeutic use , Structure-Activity Relationship , Synthetic Lethal Mutations
20.
Spectrochim Acta A Mol Biomol Spectrosc ; 316: 124326, 2024 Aug 05.
Article En | MEDLINE | ID: mdl-38669978

Based on the fact that not all chemical substances possess good Raman signals, this article focuses on the Raman silent region signals of pesticides with cyano group. Under the optimized conditions of methanol-water (1:1, v/v) as the solvent, irradiation at 302 nm light source for 20 min, and the use of 0.5 mol/L KI as the aggregating agent, Surface-enhanced Raman spectroscopy (SERS) method for azoxystrobin detection was developed by the Raman silent region signal of 2230 cm-1, and verified by detecting the spiked grapes with different concentrations of azoxystrobin. Other four pesticides with cyano group also could be identified at the peak of 2180 cm-1, 2205 cm-1, 2125 cm-1, and 2130 cm-1 for acetamiprid, phoxim, thiacloprid and cymoxanil, respectively. When azoxystrobin or acetamiprid was mixed respectively with chlorpyrifos without cyano group, their SERS signals in the Raman silent region of chlorpyrifos were not interfered, while mixed with cymoxanil in different ratios (1:4, 1:1 and 4:1), respectively, each two pesticides with cyano group could be distinguished by the changes in the Raman silent region. In further, four pesticides with or without cyano group were mixed together in 1:1:1:1 (acetamiprid, cymoxanil, azoxystrobin chlorpyrifos), and each pesticide still could be identified even at 0.5 mg/L. The results showed that the SERS method combined with UV irradiation may provide a new way to monitor the pesticides with C≡N performance in the Raman silent region without interference from the food matrix.


Pesticides , Spectrum Analysis, Raman , Strobilurins , Spectrum Analysis, Raman/methods , Pesticides/analysis , Strobilurins/analysis , Pyrimidines/analysis , Pyrimidines/chemistry , Vitis/chemistry , Methacrylates/chemistry , Methacrylates/analysis , Neonicotinoids/analysis
...