Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.002
Filter
1.
Int J Nanomedicine ; 19: 6717-6730, 2024.
Article in English | MEDLINE | ID: mdl-38979530

ABSTRACT

Introduction: Immune regulatory small molecule JQ1 can block its downstream effector PD-L1 pathway and effectively reverse the PD-L1 upregulation induced by doxorubicin (DOX). So the synergistic administration of chemotherapeutic drug DOX and JQ1 is expected to increase the sensitivity of tumors to immune checkpoint therapy and jointly enhance the body's own immunity, thus effectively killing tumor cells. Therefore, a drug delivery system loaded with DOX and JQ1 was devised in this study. Methods: Polydopamine nanoparticles (PDA NPs) were synthesized through spontaneous polymerization. Under appropriate pH conditions, DOX and JQ1 were loaded onto the surface of PDA NPs, and the release of DOX and JQ1 were measured using UV-Vis or high performance liquid chromatography (HPLC). The mechanism of fabricated nanocomplex in vitro was investigated by cell uptake experiment, cell viability assays, apoptosis assays, and Western blot analysis. Finally, the tumor-bearing mouse model was used to evaluate the tumor-inhibiting efficacy and the biosafety in vivo. Results: JQ1 and DOX were successfully loaded onto PDA NPs. PDA-DOX/JQ1 NPs inhibited the growth of prostate cancer cells, reduced the expression of apoptosis related proteins and induced apoptosis in vitro. The in vivo biodistribution indicated that PDA-DOX/JQ1 NPs could accumulated at the tumor sites through the EPR effect. In tumor-bearing mice, JQ1 delivered with PDA-DOX/JQ1 NPs reduced PD-L1 expression at tumor sites, generating significant tumor suppression. Furthermore, PDA-DOX/JQ1 NPs could reduce the side effects, and produce good synergistic treatment effect in vivo. Conclusion: We have successfully prepared a multifunctional platform for synergistic prostate cancer therapy.


Subject(s)
Apoptosis , Azepines , Doxorubicin , Indoles , Nanoparticles , Polymers , Prostatic Neoplasms , Male , Animals , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/pharmacokinetics , Doxorubicin/administration & dosage , Indoles/chemistry , Indoles/pharmacology , Indoles/pharmacokinetics , Polymers/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Nanoparticles/chemistry , Humans , Mice , Cell Line, Tumor , Apoptosis/drug effects , Azepines/chemistry , Azepines/pharmacology , Azepines/pharmacokinetics , Drug Synergism , Cell Survival/drug effects , Tissue Distribution , Xenograft Model Antitumor Assays , Drug Liberation , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , B7-H1 Antigen/metabolism , Triazoles
2.
Eur J Med Chem ; 275: 116556, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38879971

ABSTRACT

Azepanes or azepines are structural motifs of many drugs, drug candidates and evaluated lead compounds. Even though compounds having N-heterocyclic 7-membered rings are often found in nature (e.g. alkaloids), the natural compounds of this group are rather rare as approved therapeutics. Thus, recently studied and approved azepane or azepine-congeners predominantly consist of semi-synthetically or synthetically-obtained scaffolds. In this review a comparison of approved drugs and recently investigated leads was proposed taking into regard their structural aspects (stereochemistry), biological activities, pharmacokinetic properties and confirmed molecular targets. The 7-membered N-heterocycles reveal a wide range of biological activities, not only against CNS diseases, but also as e.g. antibacterial, anticancer, antiviral, antiparasitic and against allergy agents. As most of the approved or investigated potential drugs or lead structures, belonging to 7-membered N-heterocycles, are synthetic scaffolds, this report also reveals different and efficient metal-free cascade approaches useful to synthesize both simple azepane or azepine-containing congeners and those of oligocyclic structures. Stereochemistry of azepane/azepine fused systems, in view of biological data and binding with the targets, is discussed. Apart from the approved drugs, we compare advances in SAR studies of 7-membered N-heterocycles (mainly from 2018 to 2023), whereas the related synthetic part concerning various domino strategies is focused on the last ten years.


Subject(s)
Heterocyclic Compounds , Humans , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/chemical synthesis , Azepines/chemistry , Azepines/pharmacology , Azepines/chemical synthesis , Chemistry, Pharmaceutical , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Molecular Structure , Animals , Drug Approval , Stereoisomerism
3.
Adv Mater ; 36(29): e2400196, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734875

ABSTRACT

The activation of sequential events in the cancer-immunity cycle (CIC) is crucial for achieving effective antitumor immunity. However, formidable challenges, such as innate and adaptive immune resistance, along with the off-target adverse effects of nonselective immunomodulators, persist. In this study, a tumor-selective nano-regulator named PNBJQ has been presented, focusing on targeting two nonredundant immune nodes: inducing immunogenic cancer cell death and abrogating immune resistance to fully activate endogenous tumor immunity. PNBJQ is obtained by encapsulating the immunomodulating agent JQ1 within a self-assembling system formed by linking a Type-I photosensitizer to polyethylene glycol through a hypoxia-sensitive azo bond. Benefiting from the Type-I photosensitive mechanism, PNBJQ triggers the immunogenic cell death of hypoxic tumors under near-infrared (NIR) light irradiation. This process resolves innate immune resistance by stimulating sufficient cytotoxic T-lymphocytes. Simultaneously, PNBJQ smartly responds to the hypoxic tumor microenvironment for precise drug delivery, adeptly addressing adaptive immune resistance by using JQ1 to downregulate programmed death ligand 1 (PD-L1) and sustaining the response of cytotoxic T lymphocytes. The activatable synergic photoimmunotherapy promotes an immune-promoting tumor microenvironment by activating an iterative revolution of the CIC, which remarkably eradicates established hypoxic tumors and suppresses distal lesions under low light dose irradiation.


Subject(s)
Nanoparticles , Photosensitizing Agents , Animals , Mice , Cell Line, Tumor , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Nanoparticles/chemistry , Tumor Microenvironment/drug effects , Triazoles/chemistry , Triazoles/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/pathology , Immunotherapy , Azepines/chemistry , Azepines/pharmacology , Polyethylene Glycols/chemistry , Tumor Hypoxia/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/drug effects , B7-H1 Antigen/metabolism , Infrared Rays
4.
PLoS Biol ; 22(5): e3002550, 2024 May.
Article in English | MEDLINE | ID: mdl-38768083

ABSTRACT

Alkenyl oxindoles have been characterized as autophagosome-tethering compounds (ATTECs), which can target mutant huntingtin protein (mHTT) for lysosomal degradation. In order to expand the application of alkenyl oxindoles for targeted protein degradation, we designed and synthesized a series of heterobifunctional compounds by conjugating different alkenyl oxindoles with bromodomain-containing protein 4 (BRD4) inhibitor JQ1. Through structure-activity relationship study, we successfully developed JQ1-alkenyl oxindole conjugates that potently degrade BRD4. Unexpectedly, we found that these molecules degrade BRD4 through the ubiquitin-proteasome system, rather than the autophagy-lysosomal pathway. Using pooled CRISPR interference (CRISPRi) screening, we revealed that JQ1-alkenyl oxindole conjugates recruit the E3 ubiquitin ligase complex CRL4DCAF11 for substrate degradation. Furthermore, we validated the most potent heterobifunctional molecule HL435 as a promising drug-like lead compound to exert antitumor activity both in vitro and in a mouse xenograft tumor model. Our research provides new employable proteolysis targeting chimera (PROTAC) moieties for targeted protein degradation, providing new possibilities for drug discovery.


Subject(s)
Cell Cycle Proteins , Oxindoles , Proteolysis , Ubiquitin-Protein Ligases , Humans , Animals , Proteolysis/drug effects , Mice , Ubiquitin-Protein Ligases/metabolism , Oxindoles/pharmacology , Oxindoles/metabolism , Oxindoles/chemistry , Cell Cycle Proteins/metabolism , Transcription Factors/metabolism , Cell Line, Tumor , Xenograft Model Antitumor Assays , Mice, Nude , HEK293 Cells , Structure-Activity Relationship , Proteasome Endopeptidase Complex/metabolism , Azepines/pharmacology , Azepines/chemistry , Azepines/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Female , Bromodomain Containing Proteins , Receptors, Interleukin-17
5.
J Control Release ; 370: 866-878, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38685386

ABSTRACT

Glioblastoma (GBM) as one of the most lethal brain tumours, remains poor therapeutic index due to its typical characters including heterogeneous, severe immune suppression as well as the existence of blood brain barrier (BBB). Immune sonodynamic (ISD) therapy combines noninvasive sonodynamic therapy with immunotherapy, which has great prospects for the combinational treatment of GBM. Herein, we develop macrophage cell membrane cloaked reactive oxygen species (ROS) responsive biomimetic nanoparticles, co-delivering of sonosensitizer Ce6 and JQ1 (a bromo-domain protein 4 (BRD4) inhibitor which can down-regulate PD-L1) and realizing potent GBM ISD therapy. The ApoE peptide decorated macrophage membrane coating endows these biomimetic nanoparticles with low immunogenicity, efficient BBB permeability, prolonged blood circulation half-live and good biocompatibility. The ROS responsive polymeric inner core could be readily degraded as triggered by excessive ROS under the ultrasound once they accumulated in tumour cells, fast release encapsulated drugs. The generation of ROS not only killed tumour cells via sonodynamic therapy, but also induced immunogenic cell death (ICD) and further activated the anti-tumour immune response. The released JQ1 inhibited tumour cell proliferation and augmented the immune activities by inhibiting the PD-L1 expression on the surface of tumour cells. The cascade sonodynamic and immune therapy resulted in significantly improved median survival time in both orthotopic GL261 and PTEN deficient immunosuppressive CT2A GBM mice models. Therefore, our developed biomimetic nanoparticle platform provides a promising combinational therapy strategy to treat immune suppressive GBM.


Subject(s)
Brain Neoplasms , Glioblastoma , Macrophages , Nanoparticles , Reactive Oxygen Species , Triazoles , Ultrasonic Therapy , Glioblastoma/therapy , Glioblastoma/drug therapy , Glioblastoma/immunology , Animals , Ultrasonic Therapy/methods , Humans , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Brain Neoplasms/therapy , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Macrophages/drug effects , Macrophages/immunology , Nanoparticles/chemistry , Triazoles/administration & dosage , Triazoles/chemistry , Triazoles/pharmacology , Cell Membrane/metabolism , Immunotherapy/methods , Mice , Azepines/administration & dosage , Azepines/pharmacology , Azepines/chemistry , Nanomedicine/methods , Biomimetic Materials/chemistry , Female , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Blood-Brain Barrier/metabolism
6.
Curr Pharm Des ; 30(15): 1178-1193, 2024.
Article in English | MEDLINE | ID: mdl-38561613

ABSTRACT

BACKGROUND: Spinal Muscular Atrophy (SMA) is a severe motor neuronal disorder with high morbidity and mortality. Securinine has shown the potential to treat SMA; however, its anti-SMA role remains unclear. OBJECTIVE: This study aims to reveal the anti-SMA mechanisms of securinine. METHODS: Securinine-associated targets were acquired from Herbal Ingredients' Targets (HIT), Similarity Ensemble Approach (SEA), and SuperPred. SMA-associated targets were obtained from GeneCards and Dis- GeNET. Protein-protein Interaction (PPI) network was constructed using GeneMANIA, and hug targets were screened using cytoHubba. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed using ClusterProfifiler. Molecular docking was conducted using Pymol and Auto- Dock. In vitro assays were used to verify the anti-SMA effects of securinine. RESULTS: Twenty-six intersection targets of securinine and SMA were obtained. HDAC1, HDAC2, TOP2A, PIK3R1, PRMT5, JAK2, HSP90AB1, TERT, PTGS2, and PAX8 were the core targets in PPI network. GO analysis demonstrated that the intersecting targets were implicated in the regulation of proteins, steroid hormones, histone deacetylases, and DNA transcription. KEGG analysis, pathway-pathway, and hub target-pathway networks revealed that securinine might treat SMA through TNF, JAK-STAT, Ras, and PI3K-Akt pathways. Securinine had a favorable binding affinity with HDAC1, HSP90AB, JAK2, PRMT5, PTGS2, and TERT. Securinine rescued viability suppression, mitochondria damage, and SMN loss in the SMA cell model. Furthermore, securinine increased HDAC1 and PRMT5 expression, decreased PTGS2 expression, suppressed the JAK2-STAT3 pathway, and promoted the PI3K-Akt pathway. CONCLUSION: Securinine might alleviate SMA by elevating HDAC1 and PRMT5 expression and reducing PTGS2 via JAK2-STAT3 suppression and PI3K-Akt activation.


Subject(s)
Muscular Atrophy, Spinal , Network Pharmacology , Plants, Medicinal , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Humans , Plants, Medicinal/chemistry , Molecular Docking Simulation , Azepines/pharmacology , Azepines/chemistry , Azepines/isolation & purification , Lactones/pharmacology , Lactones/chemistry , Lactones/isolation & purification , Molecular Structure , Heterocyclic Compounds, Bridged-Ring , Piperidines
7.
Am J Physiol Regul Integr Comp Physiol ; 326(6): R515-R527, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38618911

ABSTRACT

Inhibition of the bromodomain and extraterminal domain (BET) protein family is a potential strategy to prevent and treat diabetes; however, the clinical use of BET bromodomain inhibitors (BETis) is associated with adverse effects. Here, we explore a strategy for targeting BETis to ß cells by exploiting the high-zinc (Zn2+) concentration in ß cells relative to other cell types. We report the synthesis of a novel, Zn2+-chelating derivative of the pan-BETi (+)-JQ1, (+)-JQ1-DPA, in which (+)-JQ1 was conjugated to dipicolyl amine (DPA). As controls, we synthesized (+)-JQ1-DBA, a non-Zn2+-chelating derivative, and (-)-JQ1-DPA, an inactive enantiomer that chelates Zn2+. Molecular modeling and biophysical assays showed that (+)-JQ1-DPA and (+)-JQ1-DBA retain potent binding to BET bromodomains in vitro. Cellular assays demonstrated (+)-JQ1-DPA attenuated NF-ĸB target gene expression in ß cells stimulated with the proinflammatory cytokine interleukin 1ß. To assess ß-cell selectivity, we isolated islets from a mouse model that expresses green fluorescent protein in insulin-positive ß cells and mTomato in insulin-negative cells (non-ß cells). Surprisingly, Zn2+ chelation did not confer ß-cell selectivity as (+)-JQ1-DPA was equally effective in both ß and α cells; however, (+)-JQ1-DPA was less effective in macrophages, a nonendocrine islet cell type. Intriguingly, the non-Zn2+-chelating derivative (+)-JQ1-DBA displayed the opposite selectivity, with greater effect in macrophages compared with (+)-JQ1-DPA, suggesting potential as a macrophage-targeting molecule. These findings suggest that Zn2+-chelating small molecules confer endocrine cell selectivity rather than ß-cell selectivity in pancreatic islets and provide valuable insights and techniques to assess Zn2+ chelation as an approach to selectively target small molecules to pancreatic ß cells.NEW & NOTEWORTHY Inhibition of BET bromodomains is a novel potential strategy to prevent and treat diabetes mellitus. However, BET inhibitors have negative side effects. We synthesized a BET inhibitor expected to exploit the high zinc concentration in ß cells to accumulate in ß cells. We show our inhibitor targeted pancreatic endocrine cells; however, it was less effective in immune cells. A control inhibitor showed the opposite effect. These findings help us understand how to target specific cells in diabetes treatment.


Subject(s)
Chelating Agents , Insulin-Secreting Cells , Zinc , Animals , Zinc/chemistry , Zinc/pharmacology , Zinc/metabolism , Chelating Agents/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mice , Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Triazoles/chemistry , Humans , Male , Azepines/pharmacology , Azepines/chemistry , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Mice, Inbred C57BL , Bromodomain Containing Proteins , Nuclear Proteins
8.
Molecules ; 29(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38675678

ABSTRACT

Bromodomain 4 and 9 (BRD4 and BRD9) have been regarded as important targets of drug designs in regard to the treatment of multiple diseases. In our current study, molecular dynamics (MD) simulations, deep learning (DL) and binding free energy calculations are integrated to probe the binding modes of three inhibitors (H1B, JQ1 and TVU) to BRD4 and BRD9. The MD trajectory-based DL successfully identify significant functional function domains, such as BC-loop and ZA-loop. The information from the post-processing analysis of MD simulations indicates that inhibitor binding highly influences the structural flexibility and dynamic behavior of BRD4 and BRD9. The results of the MM-GBSA calculations not only suggest that the binding ability of H1B, JQ1 and TVU to BRD9 are stronger than to BRD4, but they also verify that van der Walls interactions are the primary forces responsible for inhibitor binding. The hot spots of BRD4 and BRD9 revealed by residue-based free energy estimation provide target sites of drug design in regard to BRD4 and BRD9. This work is anticipated to provide useful theoretical aids for the development of selective inhibitors over BRD family members.


Subject(s)
Bromodomain Containing Proteins , Cell Cycle Proteins , Deep Learning , Molecular Dynamics Simulation , Protein Binding , Transcription Factors , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Transcription Factors/chemistry , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Humans , Binding Sites , Thermodynamics , Triazoles/chemistry , Triazoles/pharmacology , Azepines/chemistry , Azepines/pharmacology , Nuclear Proteins/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Molecular Docking Simulation
9.
Bioorg Med Chem ; 100: 117611, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38309200

ABSTRACT

Systemic inflammatory response syndrome (SIRS), an exaggerated defense response of the organism to a noxious stressor, involves a massive inflammatory cascade that ultimately leads to reversible or irreversible end-organ dysfunction and even death. Suppressing RIPK1, a key protein in necroptosis pathway, has been proven to be an effective therapeutic strategy for inflammation and SIRS. In this study, a series of novel biaryl benzoxazepinone RIPK1 inhibitors were designed and synthesized by introducing different aryl substituents at the C7 position of benzoxazepinone. As a result, p-cyanophenyl substituted analog 19 exhibited the most potent in vitro anti-necroptotic effect in HT-29 cells (EC50 = 1.7 nM) and superior protection against temperature loss and death in mice in the TZ-induced SIRS model compared to GSK'772. What's more, in vivo analysis of the levels of inflammatory factors in mice also revealed that compound 19 had better anti-inflammatory activity than GSK'772.


Subject(s)
Inflammation , Receptor-Interacting Protein Serine-Threonine Kinases , Systemic Inflammatory Response Syndrome , Animals , Humans , Mice , Apoptosis , HT29 Cells , Inflammation/metabolism , Necrosis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Systemic Inflammatory Response Syndrome/chemically induced , Systemic Inflammatory Response Syndrome/drug therapy , Systemic Inflammatory Response Syndrome/metabolism , Azepines/chemistry , Azepines/pharmacology
10.
Nucleic Acids Res ; 52(4): 1661-1676, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38084912

ABSTRACT

Bromodomain and extraterminal (BET) proteins are extensively studied in multiple pathologies, including cancer. BET proteins modulate transcription of various genes, including those synonymous with cancer, such as MYC. Thus, BET inhibitors are a major area of drug development efforts. (+)-JQ1 (JQ1) is the prototype inhibitor and is a common tool to probe BET functions. While showing therapeutic promise, JQ1 is not clinically usable, partly due to metabolic instability. Here, we show that JQ1 and the BET-inactive (-)-JQ1 are agonists of pregnane X receptor (PXR), a nuclear receptor that transcriptionally regulates genes encoding drug-metabolizing enzymes such as CYP3A4, which was previously shown to oxidize JQ1. A PXR-JQ1 co-crystal structure identified JQ1's tert-butyl moiety as a PXR anchor and explains binding by (-)-JQ1. Analogs differing at the tert-butyl lost PXR binding, validating our structural findings. Evaluation in liver cell models revealed both PXR-dependent and PXR-independent modulation of CYP3A4 expression by BET inhibitors. We have characterized a non-BET JQ1 target, a mechanism of physiological JQ1 instability, a biological function of (-)-JQ1, and BET-dependent transcriptional regulation of drug metabolism genes.


Subject(s)
Azepines , Pregnane X Receptor , Triazoles , Azepines/chemistry , Azepines/pharmacology , Cell Line, Tumor , Cell Proliferation , Cytochrome P-450 CYP3A/genetics , Nuclear Proteins/metabolism , Pregnane X Receptor/chemistry , Proto-Oncogene Proteins c-myc/genetics , Receptors, Cytoplasmic and Nuclear , Triazoles/chemistry , Triazoles/pharmacology , Humans
11.
Bioorg Chem ; 135: 106478, 2023 06.
Article in English | MEDLINE | ID: mdl-36958121

ABSTRACT

Cancer is associated with uncontrolled cell proliferation invading adjoining tissues and organs. Despite the availability of several chemotherapeutic agents, the constant search for newer approaches and drugs is necessitated owing to the ever-growing challenge of resistance. Over the years, DNA has emerged as an important druggable therapeutic drug due to its role in critical cellular processes such as cell division and maintenance. Further, evading apoptosis stands out as a hallmark of cancer. Hence, designing new compounds that would target DNA and induce apoptosis plays an important role in cancer therapy. In the current work, we carried out the synthesis and anticancer evaluation of 1-aryl-4,6-dihydrobenzo[b]pyrazolo[3,4-d]azepin-5(1H)-ones/thiones (26 compounds) against selected human cancer cell lines. Among these, compounds 8ae, 8ad, 8cf, 10ad and Kenpaullone have shown good inhibitory properties against HeLa cells (IC50 < 2 µM) with good selectivity over the non-cancerous human embryonic kidney (Hek293T) cells. In cell cycle analysis, the compounds 8ad and 8cf have exhibited G2/M cell cycle arrest in HeLa cells. In addition, the compounds 8ad and 8cf induced apoptosis in a dose-dependent manner in the Annexin-V FITC staining assay. The DAPI staining clearly demonstrated the condensed and fragmented nuclei in 8ad, 8cf, 8ae and Kenpaullone-treated HeLa cells. In addition, these compounds strongly suppressed the healing after 48 h in in vitro cell migration assay. The DNA binding experiments indicated that compounds 8ae, 8cf, and 8ad as well as Kenpaullone interact with double-stranded DNA by binding in grooves which may interrupt the DNA replication and kill fast-growing cells. Molecular docking studies revealed the binding pose of 8ad and Kenpaullone at HT1 binding pocket of double-stranded DNA. Compounds 8ad and 8cf demonstrated moderate topo II inhibition which could be a possible reason for their anticancer properties. Compounds 8ad and 8cf may cause the topo II and DNA covalent complex, which leads to the inhibition of DNA replication and transcription. This eventually increases the DNA damage in cells and promotes cell apoptosis. With the above interesting biological profile, the new 1-aryl-2,6-dihydrobenzo[b]pyrazolo[3,4-d]azepin-5(4H)-one/thione derivatives have emerged as promising leads for the discovery of new anticancer agents.


Subject(s)
Antineoplastic Agents , Thiones , Humans , Antineoplastic Agents/chemistry , Apoptosis , Cell Line, Tumor , Cell Proliferation , Drug Screening Assays, Antitumor , HEK293 Cells , HeLa Cells , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Thiones/pharmacology , Azepines/chemistry , Azepines/pharmacology
12.
Bioorg Chem ; 131: 106299, 2023 02.
Article in English | MEDLINE | ID: mdl-36493622

ABSTRACT

A series of novel substituted azepines (2-7) was synthesized using both traditional and ultrasonic techniques. The efficiency of the reaction rate and yield was improved by sonication technique. We identified the newly synthesized compounds based on their melting points, elemental analyses, and spectral data. Human cancers are regulated mainly by the phosphatidylinositol 3-kinase/protein kinases B (PI3K/Akt) pathway, and its abnormal activation is linked to carcinogenesis, and angiogenesis. Using in-silico studies, we evaluated the ability of all the novel substituted diazepines and oxazepines to prevent cancer growth and metastasis by targeting the PI3K/Akt signaling pathway. Based on our findings, compounds 4a and 7a were chosen for in-vitro testing as they ranked via molecular docking the highest binding energies of -10.9, -10.3, -10.6, and -10.4 kcal/mol respectively. Compounds 4a and 7a displayed significant cytotoxicity on Caco-2 colorectal cancer cells with IC50 values of 8.445 ± 2.26 and 33.04 ± 2.06 µM, respectively. Additionally, they considerably suppressed the PI3K/Akt proteins and generated reactive oxygen species (ROS), which increased p53 and Bax, decreased Bcl-2 levels, and arrested the cell cycle at sub-G0/G1 phase. We also observed a remarkable overexpression of the Tuberous Sclerosis Complex 2 (TSC2) gene, an inhibitor of the mammalian target of rapamycin (mTOR). These results showed that compounds 4a and 7a obeyed Lipinski's rule of five and might be potential cancer treatment scaffolds by preventing metastasis and proliferation via blocking the PI3K/Akt/TSC2/m-TOR signaling pathway. This supports our hypothesis that diazepine 4a and oxazepine 7a are promising drug candidates for colorectal cancer.


Subject(s)
Colorectal Neoplasms , Tuberous Sclerosis , Humans , Proto-Oncogene Proteins c-akt/metabolism , Sirolimus/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Molecular Docking Simulation , TOR Serine-Threonine Kinases/metabolism , Caco-2 Cells , Azepines/chemistry , Signal Transduction , Colorectal Neoplasms/metabolism , Cell Proliferation , Cell Line, Tumor
13.
Chem Pharm Bull (Tokyo) ; 70(8): 573-579, 2022.
Article in English | MEDLINE | ID: mdl-35908923

ABSTRACT

The atropisomeric properties of N-alkyl and N-aryl 4-substituted 5H-dibenz[b,f]azepines were investigated. The N-alkylation and N-arylation of 4-Cl or 4-Me substituted compounds was performed; however, none of the atropisomers produced were separated by chiral HPLC. Notably, we observed that the rotation of the four axes (ax. 1-4) in the 4-substituted 5H-dibenz[b,f]azepine structure is so rapid that N-alkylation or N-arylation is not sufficient to freeze it at room temperature. Additionally, the X-ray crystal structures of N-aryl compounds 13b and 14a indicated that the N atom in the triphenyl amine moiety in their structures shows sp2-like property.


Subject(s)
Azepines , Azepines/chemistry
14.
Molecules ; 27(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35164062

ABSTRACT

The geometry of dibenzoazepine analogues-typical multifunctional drugs-was investigated to find the geometrical parameters sensitive to the substitution of the central seven-membered ring. Exploration of the crystal structure database (CSD) shows that the geometrical parameter sensitive to the substitution of the carbon atom distance of the central ring not included in the aromatic rings to the plane through the carbon atoms common for the central ring and the aromatic side rings. Presence of the double bond in the central ring was reflected in its partial aromaticity expressed by the HOMED parameter. Some derivatives of 5H-dibenzo[b,f]azepine with flat conformation of the central ring are characterized by mobility of the electron density comparable to the mobility in the aromatic side rings. Influence of the surrounding on the investigated compounds was confirmed by comparison of the optimized molecules and the molecules in the crystal state where the packing forces can influence the molecular geometry.


Subject(s)
Azepines/chemistry , Models, Chemical , Azepines/pharmacology , Cyclization , Density Functional Theory , Molecular Structure , Thermodynamics
15.
Bioorg Med Chem ; 58: 116658, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35183880

ABSTRACT

Function-oriented molecular editing of the polycyclic scaffold of securinine led to the preparation of a library of simplified analogs that have been evaluated for their cytotoxicity potential against HCT116 and HL60 human cell lines. Chemical diversity at the C14 position (securinine numbering) was generated through the site-selective γ-iodination followed by Pd-catalyzed Sonogashira and Suzuki-Miyaura reactions. To explain the selectivity in the iodination step, a reaction mechanism has been proposed. Surprisingly, the piperidine ring (ring A) of the securinine skeleton has been found to be irrelevant for the cytotoxic activity. Based on this finding, the pharmacophoric core of securinine could be simplified to the key BCD motif. The nature of the substituent at the nitrogen can vary from a methyl or an isobutyl group to a benzyl or a carbamate moiety. Interestingly, the N-benzyl substituted simplified analog exhibited the same cytotoxic activity as the parent compound securinine. This functional group tolerance paves the way for the installation of reactive handles for the synthesis of molecular probes for target identification.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , Heterocyclic Compounds, Bridged-Ring/pharmacology , Lactones/pharmacology , Piperidines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Azepines/chemical synthesis , Azepines/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Density Functional Theory , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HCT116 Cells , HL-60 Cells , Heterocyclic Compounds, Bridged-Ring/chemical synthesis , Heterocyclic Compounds, Bridged-Ring/chemistry , Humans , Lactones/chemical synthesis , Lactones/chemistry , Molecular Conformation , Piperidines/chemical synthesis , Piperidines/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
16.
J Am Chem Soc ; 144(2): 701-708, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34994556

ABSTRACT

Proteolysis-targeting chimeras (PROTACs), heterobifunctional compounds that consist of protein-targeting ligands linked to an E3 ligase recruiter, have arisen as a powerful therapeutic modality for targeted protein degradation (TPD). Despite the popularity of TPD approaches in drug discovery, only a small number of E3 ligase recruiters are available for the >600 E3 ligases that exist in human cells. Here, we have discovered a cysteine-reactive covalent ligand, EN106, that targets FEM1B, an E3 ligase recently discovered as the critical component of the cellular response to reductive stress. By targeting C186 in FEM1B, EN106 disrupts recognition of the key reductive stress substrate of FEM1B, FNIP1. We further establish that EN106 can be used as a covalent recruiter for FEM1B in TPD applications by demonstrating that a PROTAC linking EN106 to the BET bromodomain inhibitor JQ1 or the kinase inhibitor dasatinib leads to the degradation of BRD4 and BCR-ABL, respectively. Our study showcases a covalent ligand that targets a natural E3 ligase-substrate binding site and highlights the utility of covalent ligand screening in expanding the arsenal of E3 ligase recruiters suitable for TPD applications.


Subject(s)
Acetamides/chemistry , Cell Cycle Proteins/metabolism , Proteolysis , Ubiquitin-Protein Ligase Complexes/metabolism , Animals , Azepines/chemistry , Binding Sites , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line , Cysteine/chemistry , Dasatinib/chemistry , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/metabolism , Humans , Mice , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Kinase Inhibitors/chemistry , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Triazoles/chemistry , Ubiquitin-Protein Ligase Complexes/antagonists & inhibitors , Ubiquitin-Protein Ligase Complexes/genetics
17.
Elife ; 112022 01 04.
Article in English | MEDLINE | ID: mdl-34982029

ABSTRACT

Nicotinic partial agonists provide an accepted aid for smoking cessation and thus contribute to decreasing tobacco-related disease. Improved drugs constitute a continued area of study. However, there remains no reductionist method to examine the cellular and subcellular pharmacokinetic properties of these compounds in living cells. Here, we developed new intensity-based drug-sensing fluorescent reporters (iDrugSnFRs) for the nicotinic partial agonists dianicline, cytisine, and two cytisine derivatives - 10-fluorocytisine and 9-bromo-10-ethylcytisine. We report the first atomic-scale structures of liganded periplasmic binding protein-based biosensors, accelerating development of iDrugSnFRs and also explaining the activation mechanism. The nicotinic iDrugSnFRs detect their drug partners in solution, as well as at the plasma membrane (PM) and in the endoplasmic reticulum (ER) of cell lines and mouse hippocampal neurons. At the PM, the speed of solution changes limits the growth and decay rates of the fluorescence response in almost all cases. In contrast, we found that rates of membrane crossing differ among these nicotinic drugs by >30-fold. The new nicotinic iDrugSnFRs provide insight into the real-time pharmacokinetic properties of nicotinic agonists and provide a methodology whereby iDrugSnFRs can inform both pharmaceutical neuroscience and addiction neuroscience.


Subject(s)
Alkaloids/chemistry , Azepines/chemistry , Heterocyclic Compounds, 4 or More Rings/chemistry , Nicotinic Agonists/chemistry , Smoking Cessation , Alkaloids/metabolism , Animals , Azocines/chemistry , Azocines/metabolism , Fluorescence , Humans , Ligands , Mice , Quinolizines/chemistry , Quinolizines/metabolism
18.
J Nanobiotechnology ; 19(1): 433, 2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34930269

ABSTRACT

BACKGROUND: The construction of a nanoimmune controlled-release system that spatiotemporally recognizes tumor lesions and stimulates the immune system response step by step is one of the most potent cancer treatment strategies for improving the sensitivity of immunotherapy response. RESULTS: Here, a composite nanostimulator (CNS) was constructed for the release of second near-infrared (NIR-II) photothermal-mediated immune agents, thereby achieving spatiotemporally controllable photothermal-synergized immunotherapy. CNS nanoparticles comprise thermosensitive liposomes as an outer shell and are internally loaded with a NIR-II photothermal agent, copper sulfide (CuS), toll-like receptor-9 (TLR-9) agonist, cytosine-phospho-guanine oligodeoxynucleotides, and programmed death-ligand 1 (PD-L1) inhibitors (JQ1). Following NIR-II photoirradiation, CuS enabled the rapid elevation of localized temperature, achieving tumor ablation and induction of immunogenic cell death (ICD) as well as disruption of the lipid shell, enabling the precise release of two immune-therapeutical drugs in the tumor region. Combining ICD, TLR-9 stimulation, and inhibited expression of PD-L1 allows the subsequent enhancement of dendritic cell maturation and increases infiltration of cytotoxic T lymphocytes, facilitating regional antitumor immune responses. CONCLUSION: CNS nanoparticle-mediated photothermal-synergized immunotherapy efficiently suppressed the growth of primary and distant tumors in two mouse models and prevented pulmonary metastasis. This study thus provides a novel sight into photo-controllably safe and efficient immunotherapy.


Subject(s)
Immunotherapy/methods , Infrared Rays , Nanoparticles/chemistry , Neoplasms/therapy , Phototherapy/methods , Animals , Azepines/chemistry , Azepines/pharmacology , Azepines/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Cell Line, Tumor , Copper/chemistry , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunogenic Cell Death/drug effects , Indocyanine Green/chemistry , Indocyanine Green/therapeutic use , Liposomes/chemistry , Mice , Mice, Inbred C57BL , Neoplasms/pathology , Toll-Like Receptor 9/metabolism , Transplantation, Heterologous , Triazoles/chemistry , Triazoles/pharmacology , Triazoles/therapeutic use
19.
Mar Drugs ; 19(12)2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34940713

ABSTRACT

From the marine-derived fungus Penicillium sumatrense (Trichocomaceae), a pair of enantiomers [(+)-1 and (-)-1] were isolated with identical 1D NMR data to drazepinone, which was originally reported to have a trisubstituted naphthofuroazepinone skeleton. In this study, we confirmed the structures of the two enantiomers as drazepinone and revised their structures by detailed analysis of extensive 2D NMR data and a comparison of the calculated 13C chemical shifts, ECD, VCD, and ORD spectra with those of the experiment ones. (+)-1 and (-)-1 were evaluated for their PTP inhibitory activity in vitro. (-)-1 showed selective PTP inhibitory activity against PTP1B and TCPTP with IC50 values of 1.56 and 12.5 µg/mL, respectively.


Subject(s)
Azepines/pharmacology , Enzyme Inhibitors/pharmacology , Naphthalenes/pharmacology , Penicillium , Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , Aquatic Organisms , Azepines/chemistry , Enzyme Inhibitors/chemistry , Naphthalenes/chemistry , Protein Tyrosine Phosphatases/metabolism , Structure-Activity Relationship
20.
Article in English | MEDLINE | ID: mdl-34731742

ABSTRACT

Besifloxacin has been embraced for the treatment of ocular bacterial infections. While LC-MS/MS has been used in investigating BSF pharmacokinetics, those costly instruments are not universally available and have complicated requirements for operation and maintenance. Additionally, pharmacokinetics of besifloxacin in dose-intense regimens are still unknown. Herein, a new quantification method was developed employing the widely accessible HPLC with fluorescence detection and applied to an ocular pharmacokinetic study with an intense regimen. Biosamples were pre-treated using protein precipitation. Chromatographic separation was achieved on a C18 column using mobile phase of 0.1% trifluoroacetic acid and acetonitrile. To address the weak fluorescence issue of besifloxacin, effects of detection parameters, elution pattern, pH of mobile phase, and reconstitution solvents were investigated. The method was fully validated per US-FDA guidelines and demonstrated precision (<13%), accuracy (91-112%), lower limit of quantification (5 ng/mL), linearity over clinically relevant concentrations (R2 > 0.999), matrix-effects (93-105%), recoveries (95-106%), and excellent selectivity. The method showed agreement with agar disk diffusion assays for in vitro screening and comparable in vivo performance to LC-MS/MS (Deming Regression, y = 1.010x + 0.123, r = 0.997; Bland-Altman analysis, mean difference was -6.3%; n = 21). Pharmacokinetic parameters suggested superior surface-retentive properties of besifloxacin. Maximum concentrations were 1412 ± 1910 and 0.15 ± 0.12 µg/mL; area under the curve was 1,637 and 1.08 µg·h/g; and half-life was 4.9 and 4.1 h; and pharmacokinetic-to-pharmacodynamic ratios were ≥ 409 and ≤ 17.8 against ocular pathogens in tears and aqueous humor, respectively. This readily available method is sensitive for biosamples and practical for routine use, facilitating besifloxacin therapy development.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Azepines/chemistry , Azepines/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Fluoroquinolones/chemistry , Fluoroquinolones/pharmacokinetics , Keratitis/drug therapy , Tandem Mass Spectrometry/methods , Animals , Anti-Bacterial Agents/administration & dosage , Aqueous Humor/chemistry , Azepines/administration & dosage , Chromatography, High Pressure Liquid/instrumentation , Female , Fluorescence , Fluoroquinolones/administration & dosage , Humans , Limit of Detection , Male , Rabbits , Tears/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...