Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Molecules ; 29(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38792163

ABSTRACT

To further extend the structure-activity relationships (SARs) of 5-aminopyrazoles (5APs) and identify novel compounds able to interfere with inflammation, oxidative stress, and tumorigenesis, 5APs 1-4 have been designed and prepared. Some chemical modifications have been inserted on cathecol function or in aminopyrazole central core; in detail: (i) smaller, bigger, and more lipophilic substituents were introduced in meta and para positions of catechol portion (5APs 1); (ii) a methyl group was inserted on C3 of the pyrazole scaffold (5APs 2); (iii) a more flexible alkyl chain was inserted on N1 position (5APs 3); (iv) the acylhydrazonic linker was moved from position 4 to position 3 of the pyrazole scaffold (5APs 4). All new derivatives 1-4 have been tested for radical scavenging (DPPH assay), anti-aggregating/antioxidant (in human platelets) and cell growth inhibitory activity (MTT assay) properties. In addition, in silico pharmacokinetics, drug-likeness properties, and toxicity have been calculated. 5APs 1 emerged to be promising anti-proliferative agents, able to suppress the growth of specific cancer cell lines. Furthermore, derivatives 3 remarkably inhibited ROS production in platelets and 5APs 4 showed interesting in vitro radical scavenging properties. Overall, the collected results further confirm the pharmaceutical potentials of this class of compounds and support future studies for the development of novel anti-proliferative and antioxidant agents.


Subject(s)
Antineoplastic Agents , Antioxidants , Pyrazoles , Humans , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrazoles/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antioxidants/pharmacology , Antioxidants/chemistry , Structure-Activity Relationship , Cell Line, Tumor , Cell Proliferation/drug effects , Molecular Structure
2.
ChemMedChem ; 18(17): e202300252, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37366115

ABSTRACT

In previous studies, we synthesized different imidazo-pyrazoles 1 and 2 with interesting anticancer, anti-angiogenic and anti-inflammatory activities. To further extend the structure-activity relationships of imidazo-pyrazole scaffold and to identify novel antiproliferative/anti-inflammatory agents potentially active with multi-target mechanisms, a library of compounds 3-5 has been designed and synthesized. The chemical modifications characterizing the novel derivatives include: i) decoration of the catechol ring with groups with different electronic, steric and lipophilic properties (compounds 3); ii) insertion of a methyl group on C-6 of imidazo-pyrazole scaffold (compounds 4); iii) shift of the acylhydrazonic substituent from position 7 to 6 of the imidazo-pyrazole substructure (compounds 5). All synthesized compounds were tested against a panel of cancer and normal cell lines. Derivatives 3 a, 3 e, 4 c, 5 g and 5 h showed IC50 values in the low micromolar range against selected tumor cell lines and proved to have antioxidant properties, being able to inhibit ROS production in human platelet. In silico calculation predicted favourable drug-like and pharmacokinetic properties for the most promising compounds. Furthermore, molecular docking and molecular dynamic simulations suggested the ability of most active derivative 3 e to interact with colchicine binding site in the polymeric tubulin α/tubulin ß/stathmin4 complex.


Subject(s)
Antineoplastic Agents , Tubulin , Humans , Molecular Docking Simulation , Tubulin/metabolism , Structure-Activity Relationship , Cell Line, Tumor , Pyrazoles/pharmacology , Pyrazoles/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor , Molecular Structure , Cell Proliferation
3.
ChemMedChem ; 18(9): e202300046, 2023 05 02.
Article in English | MEDLINE | ID: mdl-36806572

ABSTRACT

During the last years, we developed a large library of new selective phosphodiesterase 4D inhibitors, maintaining the catechol portion of the well-known PDE4 inhibitor Rolipram, featuring different substitutions in place of the lactam group of this reference compound. Based on the X-ray analysis of PDE4 inhibitors (PDE4Is) previously synthesized by us and of naphthyridine- and naphthyridinone-containing derivatives exhibiting PDE4 inhibitory ability described in the literature, we designed and synthesized new compounds 1-3. All of them were screened in silico as putative PDE4Is, via molecular docking studies to exploit structural variation at the catechol group to gain further contacts especially with the flat aromatic residues (Phe506 and Phe538) of enzyme. Subsequent in silico prediction of ADMET properties and in vitro biological assays on platelets and endothelial cells are in good agreement with our previous data concerning the antioxidant/anti-inflammatory activity exhibited by our previous PDE4Is and similarly to other well-known PDE4Is.


Subject(s)
Phosphodiesterase 4 Inhibitors , Phosphodiesterase 4 Inhibitors/pharmacology , Antioxidants/pharmacology , Molecular Docking Simulation , Endothelial Cells , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Catechols/chemistry
4.
Molecules ; 26(19)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34641279

ABSTRACT

(1) Background: different previously synthesized pyrazoles and imidazo-pyrazoles showed interesting anti-angiogenic action, being able to interfere with ERK1/2, AKT and p38MAPK phosphorylation in different manners and with different potency; (2) Methods: here, a new small compound library, endowed with the same differently decorated chemical scaffolds, has been synthetized to obtain new agents able to inhibit different pathways involved in inflammation, cancer and human platelet aggregation. (3) Results: most of the new synthesized derivatives resulted able to block ROS production, platelet aggregation and p38MAPK phosphorylation both in platelets and Human Umbilical Vein Endothelial cells (HUVEC). This paves the way for the development of new agents with anti-angiogenic activity.


Subject(s)
Anti-Inflammatory Agents/chemical synthesis , Imidazoles/chemistry , Pyrazoles/chemistry , Small Molecule Libraries/chemical synthesis , p38 Mitogen-Activated Protein Kinases/metabolism , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Reactive Oxygen Species/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
5.
Bioorg Chem ; 115: 105168, 2021 10.
Article in English | MEDLINE | ID: mdl-34284173

ABSTRACT

In recent years, 5-pyrazolyl-ureas have mostly been known for their attractive poly-pharmacological outline and, in particular, ethyl 1-(2-hydroxypentyl)-5-(3-(3-(trifluoromethyl) phenyl) ureido)-1H-pyrazole-4-carboxylate (named GeGe-3) has emerged as a capable anti-angiogenic compound. This paper examines its interactome by functional proteomics using a label-free mass spectrometry based platform, coupling Drug Affinity Responsive Target Stability and targeted Limited Proteolysis-Multiple Reaction Monitoring. Calreticulin has been recognized as the GeGe-3 principal target and this evidence has been supported by immunoblotting and in silico molecular docking. Furthermore, cell studies have shown that GeGe-3 lowers cell calcium mobilization, cytoskeleton organization and focal adhesion kinase expression, thus linking its biological potential to calreticulin binding and, ultimately, shedding light on the reasonable action mechanism of this molecule as an anti-angiogenic factor.


Subject(s)
Angiogenesis Inhibitors/chemistry , Proteome/metabolism , Proteomics/methods , Pyrazoles/chemistry , Urea/chemistry , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Binding Sites , Calcium/metabolism , Calreticulin/chemistry , Calreticulin/metabolism , Down-Regulation/drug effects , Focal Adhesion Kinase 1/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Ionomycin/pharmacology , Molecular Docking Simulation , Proteome/drug effects
6.
Eur J Med Chem ; 223: 113638, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34171658

ABSTRACT

Memory and cognitive functions depend on the cerebral levels of cyclic adenosine monophosphate (cAMP), which are regulated by the phosphodiesterase 4 (PDE4) family of enzymes. Selected rolipram-related PDE4 inhibitors, members of the GEBR library, have been shown to increase hippocampal cAMP levels, providing pro-cognitive benefits with a safe pharmacological profile. In a recent SAR investigation involving a subset of GEBR library compounds, we have demonstrated that, depending on length and flexibility, ligands can either adopt a twisted, an extended or a protruding conformation, the latter allowing the ligand to form stabilizing contacts with the regulatory domain of the enzyme. Here, based on those findings, we describe further chemical modifications of the protruding subset of GEBR library inhibitors and their effects on ligand conformation and potency. In particular, we demonstrate that the insertion of a methyl group in the flexible linker region connecting the catechol portion and the basic end of the molecules enhances the ability of the ligand to interact with both the catalytic and the regulatory domains of the enzyme.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Drug Design , Phosphodiesterase 4 Inhibitors/chemical synthesis , Small Molecule Libraries/chemistry , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Humans , Molecular Docking Simulation , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/metabolism , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/metabolism , Structure-Activity Relationship
7.
Molecules ; 25(15)2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32751358

ABSTRACT

The pyrazole nucleus has long been known as a privileged scaffold in the synthesis of biologically active compounds. Within the numerous pyrazole derivatives developed as potential drugs, this review is focused on molecules characterized by a urea function directly linked to the pyrazole nucleus in a different position. In the last 20 years, the interest of numerous researchers has been especially attracted by pyrazolyl-ureas showing a wide spectrum of biological activities, ranging from the antipathogenic activities (bacteria, plasmodium, toxoplasma, and others) to the anticarcinogenic activities. In particular, in the anticancer field, pyrazolyl-ureas have been shown to interact at the intracellular level on many pathways, in particular on different kinases such as Src, p38-MAPK, TrKa, and others. In addition, some of them evidenced an antiangiogenic potential that deserves to be explored. This review therefore summarizes all these biological data (from 2000 to date), including patented compounds.


Subject(s)
Chemistry, Pharmaceutical , Pharmaceutical Preparations/chemistry , Pyrazoles/chemistry , Urea/chemistry , Chemistry Techniques, Synthetic , Chemistry, Pharmaceutical/methods , Humans , Pharmaceutical Preparations/chemical synthesis , Structure-Activity Relationship
8.
ChemMedChem ; 15(11): 961-969, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32233110

ABSTRACT

Even though immunotherapy has radically changed the search for anticancer therapies, there are still many different pathways that are open to intervention with traditional small molecules. To expand our investigation in the anticancer field, we report here a new series of compounds in which our previous pyrazole and imidazopyrazole scaffolds are linked to a differently decorated phenyl ring through an acylhydrazone linker. Preliminary tests on the library were performed at the National Cancer Institute (USA) against the full NCI 60 cell panel. The best compounds among the imidazopyrazole series were then tested by immunofluorescence staining for their inhibition of cell proliferation, apoptosis induction, and their effect on the cell cycle and on microtubules. Two compounds, in particular 4-benzyloxy-3-methoxybenzyliden imidazopyrazole-7-carbohydrazide showed good growth inhibition, with IC50 values in the low-micromolar range, and induced apoptosis. Both compounds altered the cell-cycle phases with the appearance of polyploid cells. Immunofluorescence analysis evidenced microtubules alterations; tubulin polymerization assays and docking studies suggested the tubulin system to be the possible, although not exclusive, target of the new acylhydrazone series reported here.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Hydrazones/pharmacology , Microtubules/drug effects , Pyrazoles/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Hydrazones/chemical synthesis , Hydrazones/chemistry , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
9.
Molecules ; 25(4)2020 Feb 19.
Article in English | MEDLINE | ID: mdl-32093112

ABSTRACT

Alzheimer's disease is the most common type of dementia, affecting millions of people worldwide. One of its main consequences is memory loss, which is related to downstream effectors of cyclic adenosine monophosphate (cAMP). A well-established strategy to avoid cAMP degradation is the inhibition of phosphodiesterase (PDE). In recent years, GEBR-32a has been shown to possess selective inhibitory properties against PDE type 4 family members, resulting in an improvement in spatial memory processes without the typical side effects that are usually correlated with this mechanism of action. In this work, we performed the HPLC chiral resolution and absolute configuration assignment of GEBR-32a. We developed an efficient analytical and semipreparative chromatographic method exploiting an amylose-based stationary phase, we studied the chiroptical properties of both enantiomers and we assigned their absolute configuration by 1H-NMR (nuclear magnetic resonance). Lastly, we measured the IC50 values of both enantiomers against both the PDE4D catalytic domain and the long PDE4D3 isoform. Results strongly support the notion that GEBR-32a inhibits the PDE4D enzyme by interacting with both the catalytic pocket and the regulatory domains.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Phosphodiesterase 4 Inhibitors/chemistry , Humans , Nuclear Magnetic Resonance, Biomolecular
10.
Molecules ; 25(4)2020 Feb 18.
Article in English | MEDLINE | ID: mdl-32085423

ABSTRACT

Several anti-inflammatory agents based on pyrazole and imidazopyrazole scaffolds and a large library of substituted catechol PDE4D inhibitors were reported by us in the recent past. To obtain new molecules potentially able to act on different targets involved in inflammation onset we designed and synthesized a series of hybrid compounds by linking pyrazole and imidazo-pyrazole scaffolds to differently decorated catechol moieties through an acylhydrazone chain. Some compounds showed antioxidant activity, inhibiting reactive oxygen species (ROS) elevation in neutrophils, and a good inhibition of phosphodiesterases type 4D and, particularly, type 4B, the isoform most involved in inflammation. In addition, most compounds inhibited ROS production also in platelets, confirming their ability to exert an antiinflammatory response by two independent mechanism. Structure-activity relationship (SAR) analyses evidenced that both heterocyclic scaffolds (pyrazole and imidazopyrazole) and the substituted catechol moiety were determinant for the pharmacodynamic properties, even if hybrid molecules bearing to the pyrazole series were more active than the imidazopyrazole ones. In addition, the pivotal role of the catechol substituents has been analyzed. In conclusion the hybridization approach gave a new serie of multitarget antiinflammatory compounds, characterized by a strong antioxidant activity in different biological targets.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Pyrazoles/pharmacology , Reactive Oxygen Species/metabolism , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Survival/drug effects , Chemotaxis/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/pharmacology , Humans , Male , Neutrophils/drug effects , Neutrophils/metabolism , Oxidation-Reduction , Phosphodiesterase 4 Inhibitors/chemical synthesis , Phosphodiesterase 4 Inhibitors/pharmacology , Platelet Aggregation/drug effects , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...