Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.741
Filter
1.
J Agric Food Chem ; 72(19): 10772-10780, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38703122

ABSTRACT

Protoporphyrinogen IX oxidase (PPO, E.C. 1.3.3.4) plays a pivotal role in chlorophyll biosynthesis in plants, making it a prime target for herbicide development. In this study, we conducted an investigation aimed at discovering PPO-inhibiting herbicides. Through this endeavor, we successfully identified a series of novel compounds based on the pyridazinone scaffold. Following structural optimization and biological assessment, compound 10ae, known as ethyl 3-((6-fluoro-5-(6-oxo-4-(trifluoromethyl)pyridazin-1(6H)-yl)benzo[d]thiazol-2-yl)thio)propanoate, emerged as a standout performer. It exhibited robust activity against Nicotiana tabacum PPO (NtPPO) with an inhibition constant (Ki) value of 0.0338 µM. Concurrently, we employed molecular simulations to obtain further insight into the binding mechanism with NtPPO. Additionally, another compound, namely, ethyl 2-((6-fluoro-5-(5-methyl-6-oxo-4-(trifluoromethyl)pyridazin-1(6H)-yl)benzo[d]thiazol-2-yl)thio)propanoate (10bh), demonstrated broad-spectrum and highly effective herbicidal properties against all six tested weeds (Leaf mustard, Chickweed, Chenopodium serotinum, Alopecurus aequalis, Poa annua, and Polypogon fugax) at the dosage of 150 g a.i./ha through postemergence application in a greenhouse. This work identified a novel lead compound (10bh) that showed good activity in vitro and excellent herbicidal activity in vivo and had promising prospects as a new PPO-inhibiting herbicide lead.


Subject(s)
Drug Design , Enzyme Inhibitors , Herbicides , Nicotiana , Plant Proteins , Protoporphyrinogen Oxidase , Pyridazines , Protoporphyrinogen Oxidase/antagonists & inhibitors , Protoporphyrinogen Oxidase/metabolism , Protoporphyrinogen Oxidase/chemistry , Protoporphyrinogen Oxidase/genetics , Pyridazines/chemistry , Pyridazines/pharmacology , Herbicides/pharmacology , Herbicides/chemistry , Herbicides/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemical synthesis , Structure-Activity Relationship , Nicotiana/metabolism , Nicotiana/enzymology , Plant Proteins/chemistry , Plant Proteins/metabolism , Plant Proteins/antagonists & inhibitors , Plant Proteins/genetics , Molecular Docking Simulation , Molecular Structure , Plant Weeds/drug effects , Plant Weeds/enzymology , Kinetics
2.
Sci Rep ; 14(1): 10689, 2024 05 09.
Article in English | MEDLINE | ID: mdl-38724641

ABSTRACT

Homeostatic plasticity, the ability of neurons to maintain their averaged activity constant around a set point value, is thought to account for the central hyperactivity after hearing loss. Here, we investigated the putative role of GABAergic neurotransmission in this mechanism after a noise-induced hearing loss larger than 50 dB in high frequencies in guinea pigs. The effect of GABAergic inhibition is linked to the normal functioning of K + -Cl- co-transporter isoform 2 (KCC2) which maintains a low intracellular concentration of chloride. The expression of membrane KCC2 were investigated before and after noise trauma in the ventral and dorsal cochlear nucleus (VCN and DCN, respectively) and in the inferior colliculus (IC). Moreover, the effect of gabazine (GBZ), a GABA antagonist, was also studied on the neural activity in IC. We show that KCC2 is downregulated in VCN, DCN and IC 3 days after noise trauma, and in DCN and IC 30 days after the trauma. As expected, GBZ application in the IC of control animals resulted in an increase of spontaneous and stimulus-evoked activity. In the noise exposed animals, on the other hand, GBZ application decreased the stimulus-evoked activity in IC neurons. The functional implications of these central changes are discussed.


Subject(s)
Hearing Loss, Noise-Induced , K Cl- Cotransporters , Symporters , gamma-Aminobutyric Acid , Animals , Symporters/metabolism , Symporters/antagonists & inhibitors , Guinea Pigs , Hearing Loss, Noise-Induced/metabolism , Hearing Loss, Noise-Induced/physiopathology , gamma-Aminobutyric Acid/metabolism , Male , Cochlear Nucleus/metabolism , Pyridazines/pharmacology , Neurons/metabolism
3.
Pestic Biochem Physiol ; 201: 105888, 2024 May.
Article in English | MEDLINE | ID: mdl-38685219

ABSTRACT

Bemisia tabaci is a formidable insect pest worldwide, and it exhibits significant resistance to various insecticides. Dimpropyridaz is a novel pyridazine pyrazolecarboxamide insecticide used against sucking insect pests, but there is little information regarding its metabolic detoxification in arthropods or cross-resistance with other insecticides. In this study, we found that dimpropyridaz shows no cross-resistance with three other popular insecticides, namely abamectin, cyantraniliprole, and flupyradifurone. After treatment of B. tabaci adults with a high dose of dimpropyridaz, higher cytochrome P450 monooxygenase (P450) activity was detected in the survivors, and the expression of the P450 gene CYP6DW4 was highly induced. Cloning and characterization of the full-length amino acid sequence of CYP6DW4 indicated that it contains conserved domains typical of P450 genes, phylogenetic analysis revealed that it was closely related to a B. tabaci protein, CYP6DW3, known to be involved in detoxification of imidacloprid. Silencing of CYP6DW4 by feeding insects with dsRNA significantly increased the susceptibility of B. tabaci to dimpropyridaz. In addition, homology modeling and molecular docking analyses showed the stable binding of dimpropyridaz to CYP6DW4, with binding free energy of -6.65 kcal/mol. Our findings indicate that CYP6DW4 plays an important role in detoxification of dimpropyridaz and possibly promotes development of resistance in B. tabaci.


Subject(s)
Cytochrome P-450 Enzyme System , Hemiptera , Insect Proteins , Insecticide Resistance , Insecticides , Ivermectin/analogs & derivatives , Pyrazoles , Pyridazines , ortho-Aminobenzoates , Animals , Hemiptera/drug effects , Hemiptera/genetics , Insecticides/pharmacology , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Pyridazines/pharmacology , Insecticide Resistance/genetics , Insect Proteins/genetics , Insect Proteins/metabolism , Insect Proteins/chemistry , Pyrazoles/pharmacology , Phylogeny , Neonicotinoids/pharmacology , Gene Knockdown Techniques , Molecular Docking Simulation , Amino Acid Sequence , Ivermectin/pharmacology , Ivermectin/toxicity
4.
Drug Dev Res ; 85(2): e22173, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38515272

ABSTRACT

New pyridazine and pyridazinone derivatives 3a-g, 4a-f, 6a, and 6b were designed and synthesized. Cell viability of all compounds was established based on the viability of lipopolysaccharide-induced RAW264.7 macrophage cells determined via the MTT assay. In vitro inhibition assays on human COX-1 and COX-2 enzymes were conducted to probe the newly synthesized compounds' anti-inflammatory activity. The half maximal inhibitory concentration values for the most active compounds, 3d, 3e, and 4e towards COX-2 were 0.425, 0.519, and 0.356 µM, respectively, in comparison with celecoxib. The newly synthesized compounds' ability to inhibit the production of certain proinflammatory cytokines, such as inducible nitric oxide synthase, tumor necrosis factor-α, interleukin-6, and prostaglandin-E2, was also estimated in lipopolysaccharide-induced macrophages (RAW264.7 cells). Compounds 3d and 3e were identified as the most potent cytokine production inhibitors. The results of molecular modeling studies suggested that these compounds were characterized by a reasonable binding affinity toward the active site of COX-2, when compared to a reference ligand. These results might be taken into consideration in further investigations into new anti-inflammatory agents.


Subject(s)
Lipopolysaccharides , Pyridazines , Mice , Animals , Humans , Lipopolysaccharides/pharmacology , Cyclooxygenase 2/metabolism , Macrophages/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/metabolism , RAW 264.7 Cells , Pyridazines/pharmacology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism
5.
Chem Biodivers ; 21(5): e202400110, 2024 May.
Article in English | MEDLINE | ID: mdl-38424689

ABSTRACT

Drugs with anti-platelet aggregation and neuroprotection are of great significance for the treatment of ischemic stroke. A series of edaravone and 6-phenyl-4,5-dihydropyridazin-3(2H)-one hybrids were designed and synthesized. Among them, 6g showed the most effective cytoprotective effect against oxygen-glucose deprivation/reoxygenation-induced damage in BV2 cells and an excellent inhibitory effect on platelet aggregation induced by adenosine diphosphate and arachidonic acid. Additionally, 6g could prevent thrombosis caused by ferric chloride in rats and pose a lower risk of causing bleeding compared with aspirin. It provides better protection against ischemia/reperfusion injury in rats compared with edaravone and alleviates the oxidative stress related to cerebral ischemia/reperfusion by increasing the GSH and SOD levels and decreasing the MDA concentration. Finally, molecular docking results showed that 6g probably acts on PDE3 A and plays an anti-platelet aggregation effect. Overall, 6g could be a potential candidate compound for the treatment of ischemic stroke.


Subject(s)
Edaravone , Ischemic Stroke , Neuroprotective Agents , Platelet Aggregation Inhibitors , Platelet Aggregation , Animals , Edaravone/pharmacology , Edaravone/chemistry , Ischemic Stroke/drug therapy , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Rats , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/chemical synthesis , Platelet Aggregation/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/chemical synthesis , Molecular Docking Simulation , Male , Mice , Molecular Structure , Structure-Activity Relationship , Rats, Sprague-Dawley , Drug Discovery , Pyridazines/pharmacology , Pyridazines/chemistry , Oxidative Stress/drug effects
7.
Leukemia ; 38(3): 475-481, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38287132

ABSTRACT

Ponatinib, the only approved all known-BCR::ABL1 inhibitor, is a third-generation tyrosine-kinase inhibitor (TKI) designed to inhibit BCR::ABL1 with or without any single resistance mutation, including T315I, and induced robust and durable responses at 45 mg/day in patients with CP-CML resistant to second-generation TKIs in the PACE trial. However, cardiovascular toxicities, including arterial occlusive events (AOEs), have emerged as treatment-related AEs within this class of TKIs. The OPTIC trial evaluated the efficacy and safety of ponatinib using a novel, response-based, dose-reduction strategy in patients with CP-CML whose disease is resistant to ≥2 TKIs or who harbor T315I. To assess the dose-response relationship and the effect on the safety of ponatinib, we examined the outcomes of patients with CP-CML enrolled in PACE and OPTIC who received 45 mg/day of ponatinib. A propensity score analysis was used to evaluate AOEs across both trials. Survival rates and median time to achieve ≤1% BCR::ABL1IS in OPTIC were similar or better than in PACE. The outcomes of patients with T315I mutations were robust in both trials. Patients in OPTIC had a lower exposure-adjusted incidence of AOEs compared with those in PACE. This analysis demonstrates that response-based dosing for ponatinib improves treatment tolerance and mitigates cardiovascular risk.


Subject(s)
Antineoplastic Agents , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Leukemia, Myeloid, Chronic-Phase , Pyridazines , Humans , Drug Resistance, Neoplasm , Leukemia, Myeloid, Chronic-Phase/drug therapy , Leukemia, Myeloid, Chronic-Phase/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Imidazoles/therapeutic use , Imidazoles/pharmacology , Pyridazines/therapeutic use , Pyridazines/pharmacology , Fusion Proteins, bcr-abl/genetics , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
8.
J Biomol Struct Dyn ; 42(5): 2242-2256, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37211823

ABSTRACT

Developing highly potent covalent inhibitors of Fibroblast growth factor receptors 1 (FGFR1) has always been a challenging task. In the current study, various computational techniques, such as 3D-QSAR, covalent docking, fingerprinting analysis, MD simulation followed by MMGB/PBSA, and per-residue energy decomposition analysis were used to explore the binding mechanism of pyrazolo[3,4-d]pyridazinone derivatives to FGFR1. The high q2 and r2 values for the CoMFA and CoMSIA models, suggest that the constructed 3D-QSAR models could reliably predict the bioactivities of FGFR1 inhibitors. The structural requirements revealed by the model's contour maps were strategically used to computationally create an in-house library of more than 100 new FGFR1 inhibitors using the R-group exploration technique implemented in the SparkTM software. The compounds from the in-house library were also mapped in the 3D-QSAR model that predicts comparable pIC50 values with the experimental values. A comparison between 3D-QSAR generated contours and molecular docking conformation of ligands was performed to reveal the fundamentals to design potent FGFR1 covalent inhibitors. The estimated binding free energies (MMGB/PBSA) for the selected compounds were in agreement with the experimental value ranking of their binding affinities towards FGFR1. Furthermore, per-residue energy decomposition analysis has identified Arg627 and Glu531 to contribute significantly in improved binding affinity of compound W16. During ADME analysis, the majority of in-house library compounds exhibited pharmacokinetic properties superior to those of experimentally produced compounds. These new compounds may help researchers better understand FGFR1 inhibition and lead to the creation of novel, potent FGFR1 inhibitors.Communicated by Ramaswamy H. Sarma.


Subject(s)
Antineoplastic Agents , Molecular Dynamics Simulation , Pyrazoles , Pyridazines , Receptor, Fibroblast Growth Factor, Type 1 , Antineoplastic Agents/pharmacology , Molecular Docking Simulation , Protein Binding , Quantitative Structure-Activity Relationship , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyridazines/chemistry , Pyridazines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
9.
J Biol Inorg Chem ; 29(1): 51-73, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38099936

ABSTRACT

Platinum-based drugs are widely recognized efficient anti-tumor agents, but faced with multiple undesirable effects. Here, four dinuclear platinum(II) complexes, [{Pt(1,2-pn)Cl}2(µ-pydz)]Cl2 (C1), [{Pt(ibn)Cl}2(µ-pydz)]Cl2 (C2), [{Pt(1,3-pn)Cl}2(µ-pydz)]Cl2 (C3) and [{Pt(1,3-pnd)Cl}2(µ-pydz)]Cl2 (C4), were designed (pydz is pyridazine, 1,2-pn is ( ±)-1,2-propylenediamine, ibn is 1,2-diamino-2-methylpropane, 1,3-pn is 1,3-propylenediamine, and 1,3-pnd is 1,3-pentanediamine). Interactions and binding ability of C1-C4 complexes with calf thymus DNA (CT-DNA) has been monitored by viscosity measurements, UV-Vis, fluorescence emission spectroscopy and molecular docking. Binding affinities of C1-C4 complexes to the bovine serum albumin (BSA) has been monitored by fluorescence emission spectroscopy. The tested complexes exhibit variable cytotoxicity toward different mouse and human tumor cell lines. C2 shows the most potent cytotoxicity, especially against mouse (4T1) and human (MDA-MD468) breast cancer cells in the dose- and time-dependent manner. C2 induces 4T1 and MDA-MD468 cells apoptosis, further documented by the accumulation of cells at sub-G1 phase of cell cycle and increase of executive caspase 3 and caspase 9 levels in 4T1 cells. C2 exhibits anti-proliferative effect through the reduction of cyclin D3 and cyclin E expression and elevation of inhibitor p27 level. Also, C2 downregulates c-Myc and phosphorylated AKT, oncogenes involved in the control of tumor cell proliferation and death. In order to measure the amount of platinum(II) complexes taken up by the cells, the cellular platinum content were quantified. However, C2 failed to inhibit mouse breast cancer growth in vivo. Chemical modifications of tested platinum(II) complexes might be a valuable approach for the improvement of their anti-tumor activity, especially effects in vivo.


Subject(s)
Antineoplastic Agents , Breast Neoplasms , Coordination Complexes , Pyridazines , Humans , Animals , Mice , Female , Platinum/pharmacology , Platinum/chemistry , Serum Albumin, Bovine/chemistry , Molecular Docking Simulation , Ligands , DNA/chemistry , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Pyridazines/pharmacology , Coordination Complexes/pharmacology , Coordination Complexes/chemistry
10.
Molecules ; 28(21)2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37959672

ABSTRACT

The strategic planning of this study is based upon using the nanoformulation method to prepare nanoparticles 4-SLNs and 4-LPHNPs of the previously prepared 4,5-diphenyl-1H-pyrazolo[3,4-c]pyridazin-3-amine (4) after confirming its structure with single crystal X-ray analysis. These nanoparticles exhibited promising cytotoxic activity against HepG-2, HCT-116 and MCF-7 cancer cell lines in comparison with the reference doxorubicin and the original derivative 4. Moreover, their inhibitory assessment against EGFR and CDK-2/cyclin A2 displayed improved and more favorable impact than the parent 4 and the references. Detection of their influence upon cancer biomarkers revealed upregulation of Bax, p53 and caspase-3 levels and downregulation of Bcl-2 levels. The docking simulation demonstrated that the presence of the pyrazolo[3,4-c]pyridazin-3-amine scaffold is amenable to enclosure and binding well within EGFR and CDK-2 receptors through different hydrophilic interactions. The pharmacokinetic and physicochemical properties of target 4 were also assessed with ADME investigation, and the outcome indicated good drug-like characteristics.


Subject(s)
Antineoplastic Agents , Nanoparticles , Pyridazines , Humans , Structure-Activity Relationship , Cell Proliferation , Cell Line, Tumor , Antineoplastic Agents/chemistry , ErbB Receptors/metabolism , Pyridazines/pharmacology , Amines/pharmacology , Molecular Structure , Molecular Docking Simulation , Drug Screening Assays, Antitumor , Protein Kinase Inhibitors/chemistry
11.
Ital J Pediatr ; 49(1): 141, 2023 Oct 15.
Article in English | MEDLINE | ID: mdl-37840126

ABSTRACT

BACKGROUND: The hemodynamic status of newborns with intracranial arteriovenous shunts (AVSs) may be extremely complex. Mini-invasive hemodynamic monitoring through innovative techniques such as Near-Infrared Spectroscopy (NIRS) and Pressure Recording Analytical Method (PRAM) may help in understanding hemodynamics in newborns with AVSs. Levosimendan is a calcium sensitizer and inodilator, and it is known to improve ventricular function, but its use in newborns is limited. In our cases, we evaluated the effect of levosimendan on hemodynamics through NIRS and PRAM. CASE PRESENTATION: Herein, we report the cases of two neonates with intracranial arteriovenous shunts, in whom we used levosimendan to manage cardiac failure refractory to conventional treatment. Levosimendan was used at a dosage of 0.1 mcg/kg/min for 72 h. Combined use of NIRS and PRAM helped in real-time monitoring of hemodynamic effects; in particular, levosimendan determined significant improvement in myocardium contractility as well as a reduction of heart rate. CONCLUSION: In two neonatal cases of AVSs, levosimendan led to an overall hemodynamic stabilization, documented by the combination of NIRS and PRAM. Our results suggest introducing levosimendan as a second-line treatment in cases of severe cardiac dysfunction due to AVSs without improvement using standard treatment strategies. Future prospective and larger studies are highly warranted.


Subject(s)
Heart Failure , Pyridazines , Humans , Infant, Newborn , Simendan/pharmacology , Cardiotonic Agents/therapeutic use , Cardiotonic Agents/pharmacology , Hydrazones/therapeutic use , Hydrazones/pharmacology , Pyridazines/therapeutic use , Pyridazines/pharmacology , Heart Failure/drug therapy , Hemodynamics
12.
Scand J Trauma Resusc Emerg Med ; 31(1): 61, 2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37880801

ABSTRACT

BACKGROUND: Accidental hypothermia, recognized by core temperature below 35 °C, is a lethal condition with a mortality rate up to 25%. Hypothermia-induced cardiac dysfunction causing increased total peripheral resistance and reduced cardiac output contributes to the high mortality rate in this patient group. Recent studies, in vivo and in vitro, have suggested levosimendan, milrinone and isoprenaline as inotropic treatment strategies in this patient group. However, these drugs may pose increased risk of ventricular arrhythmias during hypothermia. Our aim was therefore to describe the effects of levosimendan, milrinone and isoprenaline on the action potential in human cardiomyocytes during hypothermia. METHODS: Using an experimental in vitro-design, levosimendan, milrinone and isoprenaline were incubated with iCell2 hiPSC-derived cardiomyocytes and cellular action potential waveforms and contraction were recorded from monolayers of cultured cells. Experiments were conducted at temperatures from 37 °C down to 26 °C. One-way repeated measures ANOVA was performed to evaluate differences from baseline recordings and one-way ANOVA was performed to evaluate differences between drugs, untreated control and between drug concentrations at the specific temperatures. RESULTS: Milrinone and isoprenaline both significantly increases action potential triangulation during hypothermia, and thereby the risk of ventricular arrhythmias. Levosimendan, however, does not increase triangulation and the contractile properties also remain preserved during hypothermia down to 26 °C. CONCLUSIONS: Levosimendan remains a promising candidate drug for inotropic treatment of hypothermic patients as it possesses ability to treat hypothermia-induced cardiac dysfunction and no increased risk of ventricular arrhythmias is detected. Milrinone and isoprenaline, on the other hand, appears more dangerous in the hypothermic setting.


Subject(s)
Heart Diseases , Hypothermia , Pyridazines , Humans , Simendan , Milrinone/pharmacology , Milrinone/therapeutic use , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Isoproterenol/pharmacology , Hypothermia/chemically induced , Myocytes, Cardiac , Hydrazones/pharmacology , Hydrazones/therapeutic use , Pyridazines/pharmacology , Pyridazines/therapeutic use , Heart Diseases/drug therapy
13.
Altern Ther Health Med ; 29(8): 668-673, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37678873

ABSTRACT

Objective: To systematically evaluate the effect of levosimendan on cardiac function and outcomes in patients with sepsis. Method: We searched multiple databases including CNKI, VIP, WanFang Data, WOS, PubMed, EMbase, and The Cochrane Library up to February 2023. We targeted RCTs comparing levosimendan with dobutamine as a control for treating sepsis. After a rigorous screening and quality evaluation, 18 studies were selected for meta-analysis using Review Manager 5.4. Results: Out of 18 studies involving 980 sepsis patients, the meta-analysis revealed the following for the levosimendan group compared to dobutamine: (1) A significant reduction in mortality rate (OR = 0.63, 95% CI (0.42,0.95), P = .03). (2) Shortened ICU stay (MD = -2.55, 95% CI (-3.12, -1.98), P < .00001). (3) Increased left ventricular ejection fraction (LVEF) (MD = 6.05, 95%CI (5.28, 6.81), P < .00001) and cardiac index (CI) (MD = 0.47, 95%CI (0.35, 0.59), P < .00001). (4) Decreased blood lactate (Lac) (MD = -1.31, 95%CI (-1.73, -0.90), P < .00001) and troponin I (TnI) levels (MD = -0.43, 95%CI (-0.66, -0.21), P = .0002). (5) Reduced incidence of adverse events (OR = 0.43, 95% CI (0.23,0.81), P = .008). Conclusions: Compared to dobutamine, levosimendan substantially enhances cardiac function in sepsis patients, leading to improved outcomes and fewer adverse events.


Subject(s)
Pyridazines , Sepsis , Shock, Septic , Humans , Simendan/therapeutic use , Dobutamine/pharmacology , Dobutamine/therapeutic use , Stroke Volume , Hydrazones/pharmacology , Pyridazines/pharmacology , Ventricular Function, Left , Sepsis/drug therapy
14.
J Med Chem ; 66(16): 11133-11157, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37535845

ABSTRACT

FLT3 kinase is a potential drug target in acute myeloid leukemia (AML). Patients with FLT3 mutations typically have higher relapse rates and worse outcomes than patients without FLT3 mutations. In this study, we investigated the suitability of various heterocycles as central cores of FLT3 inhibitors, including thieno[3,2-d]pyrimidine, pyrazolo[1,5-a]pyrimidine, imidazo[4,5-b]pyridine, pyrido[4,3-d]pyrimidine, and imidazo[1,2-b]pyridazine. Our assays revealed a series of imidazo[1,2-b]pyridazines with high potency against FLT3. Compound 34f showed nanomolar inhibitory activity against recombinant FLT3-ITD and FLT3-D835Y (IC50 values 4 and 1 nM, respectively) as well as in the FLT3-ITD-positive AML cell lines MV4-11, MOLM-13, and MOLM-13 expressing the FLT3-ITD-D835Y mutant (GI50 values of 7, 9, and 4 nM, respectively). In contrast, FLT3-independent cell lines were much less sensitive. In vitro experiments confirmed suppression of FLT3 downstream signaling pathways. Finally, the treatment of MV4-11 xenograft-bearing mice with 34f at doses of 5 and 10 mg/kg markedly blocked tumor growth without any adverse effects.


Subject(s)
Antineoplastic Agents , Leukemia, Myeloid, Acute , Pyridazines , Humans , Mice , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridazines/pharmacology , Pyridazines/therapeutic use , Leukemia, Myeloid, Acute/pathology , Pyrimidines/pharmacology , fms-Like Tyrosine Kinase 3/genetics , Mutation , Apoptosis
15.
Molecules ; 28(14)2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37513351

ABSTRACT

Secure and efficient treatment of diverse pain and inflammatory disorders is continually challenging. Although NSAIDs and other painkillers are well-known and commonly available, they are sometimes insufficient and can cause dangerous adverse effects. As yet reported, derivatives of pyrrolo[3,4-d]pyridazinone are potent COX-2 inhibitors with a COX-2/COX-1 selectivity index better than meloxicam. Considering that N-acylhydrazone (NAH) moiety is a privileged structure occurring in many promising drug candidates, we decided to introduce this pharmacophore into new series of pyrrolo[3,4-d]pyridazinone derivatives. The current paper presents the synthesis and in vitro, spectroscopic, and in silico studies evaluating the biological and physicochemical properties of NAH derivatives of pyrrolo[3,4-d]pyridazinone. Novel compounds 5a-c-7a-c were received with high purity and good yields and did not show cytotoxicity in the MTT assay. Their COX-1, COX-2, and 15-LOX inhibitory activities were estimated using enzymatic tests and molecular docking studies. The title N-acylhydrazones appeared to be promising dual COX/LOX inhibitors. Moreover, spectroscopic and computational methods revealed that new compounds form stable complexes with the most abundant plasma proteins-AAG and HSA, but do not destabilize their secondary structure. Additionally, predicted pharmacokinetic and drug-likeness properties of investigated molecules suggest their potentially good membrane permeability and satisfactory bioavailability.


Subject(s)
Cyclooxygenase Inhibitors , Hydrazones , Lipoxygenase Inhibitors , Pyridazines , Pyrroles , Hydrazones/chemical synthesis , Hydrazones/chemistry , Hydrazones/pharmacokinetics , Hydrazones/pharmacology , Cyclooxygenase Inhibitors/chemical synthesis , Cyclooxygenase Inhibitors/chemistry , Cyclooxygenase Inhibitors/pharmacokinetics , Cyclooxygenase Inhibitors/pharmacology , Pyridazines/chemical synthesis , Pyridazines/chemistry , Pyridazines/pharmacokinetics , Pyridazines/pharmacology , Pyrroles/chemical synthesis , Pyrroles/chemistry , Pyrroles/pharmacokinetics , Pyrroles/pharmacology , Humans , Fibroblasts , Computer Simulation , Cell Membrane Permeability , Cell Line
16.
Bioorg Med Chem ; 88-89: 117332, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37210791

ABSTRACT

Pyridachlometyl is a unique pyridazine fungicide with a novel mode of action. Herein, we describe the pathway for the invention of pyridachlometyl. First, we identified a diphenyl-imidazo[1,2-a]pyrimidine as our proprietary lead with potent fungicidal activity. Then, aiming to simplify the chemical structure, we applied judicious estimations to explore monocyclic heterocycles as pharmacophores. This enabled the identification of a novel class of tetrasubstituted pyridazine compounds with potent fungicidal activity, likely retaining the same mode of action as the aforementioned compounds. The findings indicated bioisosteric similarity between diphenyl-imidazo[1,2-a]pyrimidine and pyridazine. Further structure-activity and mammalian safety investigations of pyridazine compounds resulted in the discovery of pyridachlometyl as a candidate for commercial development.


Subject(s)
Fungicides, Industrial , Pyridazines , Animals , Fungicides, Industrial/pharmacology , Fungicides, Industrial/chemistry , Biphenyl Compounds , Pyrimidines/pharmacology , Pyridazines/pharmacology , Pyridazines/chemistry , Structure-Activity Relationship , Mammals
17.
Leukemia ; 37(6): 1194-1203, 2023 06.
Article in English | MEDLINE | ID: mdl-37076694

ABSTRACT

LCK is a novel therapeutic target in ~40% of T-cell acute lymphoblastic leukemia (T-ALL), and dasatinib and ponatinib can act as LCK inhibitors with therapeutic effects. We herein report a comprehensive preclinical pharmacokinetic and pharmacodynamic evaluation of dasatinib and ponatinib in LCK-activated T-ALL. In 51 human T-ALL cases, these two drugs showed similar patterns of cytotoxic activity, with ponatinib being slightly more potent. Given orally in mice, ponatinib was associated with slower clearance with a longer Tmax and higher AUC0-24 h, although maximum pLCK inhibition was comparable between the two drugs. After establishing the exposure-to-response models, we simulated the steady-state pLCK inhibitory effects of each drug at currently approved dosages in humans: dasatinib at 140 mg and ponatinib at 45 mg once daily are both sufficient to achieve >50% pLCK inhibition for 13.0 and 13.9 h/day, respectively, comparable to pharmacodynamic profiles of these agents in BCR::ABL1 leukemias. Moreover, we developed a dasatinib-resistant T-ALL cell line model with LCK T316I mutation, in which ponatinib retained partial activity against LCK. In conclusion, we described the pharmacokinetic and pharmacodynamic profiles of dasatinib and ponatinib as LCK inhibitors in T-ALL, providing critical data for the development of human trials of these agents.


Subject(s)
Antineoplastic Agents , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Pyridazines , Humans , Animals , Mice , Dasatinib/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Pyridazines/pharmacology , Pyridazines/therapeutic use , T-Lymphocytes/metabolism , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism
18.
Top Companion Anim Med ; 53-54: 100769, 2023.
Article in English | MEDLINE | ID: mdl-36813230

ABSTRACT

This study aimed to investigate the effects of intravenous pimobendan on cardiovascular function and to determine the appropriate dose for clinical usage in cats. Six purpose-bred cats received one of the following treatments: intravenous pimobendan at a single dose of 0.075 mg/kg (low dose [LD] group), 0.15 mg/kg (middle dose [MD] group), 0.3 mg/kg (high dose [HD] group), or saline at 0.1 mL/kg (placebo group). Echocardiography and blood pressure measurements were performed before and 5, 15, 30, 45, and 60 minute after drug administration for each treatment. In the MD and HD groups, the fractional shortening, peak systolic velocity, cardiac output, and heart rate increased significantly. There were no significant differences in blood pressure among the groups. Intravenous pimobendan at 0.15-0.3 mg/kg increased the fractional shortening, peak systolic velocity, cardiac output in healthy cats.


Subject(s)
Cardiotonic Agents , Pyridazines , Cats , Animals , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Pyridazines/pharmacology , Pyridazines/therapeutic use , Blood Pressure , Echocardiography/veterinary
19.
Mol Divers ; 27(6): 2729-2740, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36534357

ABSTRACT

A series of novel pyridazine-acetohydrazide hybrids were designed, synthesized, and evaluated for their in vitro and in vivo antihyperglycemic activity. In this context, pyridazine-acetohydrazides (6a-6p) were synthesized by coupling substituted aldehyde with 2-(5-cyano-6-oxo-3,4-diphenylpyridazine-1-6H-yl) acetohydrazide, which was prepared via the reaction of pyridazine ester with hydrazine hydrate. The molecular docking study was carried out to examine the binding affinities and interaction of designed compounds against the DPP-4 enzyme. Compounds 6e, 6f, 6l, and 6n exhibited interaction with active residue. In silico ADMET properties, and toxicity studies corroborated that compounds were found to have good bioavailability and less toxic. The synthesized compounds were further estimated for in vitro DPP-4 activity. Compounds 6e and 6l were found as the most effective DPP-4 inhibitor in this series with IC50 values (6.48, 8.22 nM) when compared with sitagliptin (13.02 nM). According to the toxicity assay compound, 6l showed very less toxicity at a higher concentration so further selected for the in vivo antihyperglycemic activity.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors , Pyridazines , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Structure-Activity Relationship , Molecular Docking Simulation , Ligands , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/chemistry , Pyridazines/pharmacology
20.
Acta Cardiol ; 78(2): 170-187, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36222590

ABSTRACT

Inotropic agents are generally recommended to use in patients with acute decompensated heart failure (HF) with reduced ejection fraction (HFrEF) concurrent to end-organ dysfunction. However, due to certain pharmacological limitations like developing life threatening arrhythmia and tolerance, cannot be employed as much as needed. Meanwhile, Calcium ion (Ca2+) sensitisers exhibits their inotropic action by increasing the sensitivity of the cardiomyocyte to intracellular Ca2+ ion and have been reported as emerging therapeutic alternative in HF cases. Levosimendan (LEVO) is an inodilator and with its unique pharmacology justifying its use in a wide range of cardiac alterations in HF particularly in undergoing cardiac surgery. It is also reported to be better than classical inotropes in maintaining cardiac mechanical efficacy and reducing congestion in acute HF with hypotension. This review paper was designed to compile various evidence about basic pharmacology and potential clinical aspects of LEVO in cardiac surgery and other HF associated alterations. This will benefit directly to the researcher in initiating research and to fill the gaps in the area of thrust.


Subject(s)
Heart Failure , Pyridazines , Humans , Simendan/pharmacology , Simendan/therapeutic use , Heart Failure/drug therapy , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Hydrazones/pharmacology , Hydrazones/therapeutic use , Pyridazines/pharmacology , Pyridazines/therapeutic use , Stroke Volume , Myocytes, Cardiac
SELECTION OF CITATIONS
SEARCH DETAIL
...