Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Bioorg Med Chem ; 24(14): 3174-83, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27265687

ABSTRACT

The clonidine-like central antihypertensive agent rilmenidine, which has high affinity for I1-type imidazoline receptors (I1-IR) was recently found to have cytotoxic effects on cultured cancer cell lines. However, due to its pharmacological effects resulting also from α2-adrenoceptor activation, rilmenidine cannot be considered a suitable anticancer drug candidate. Here, we report the identification of novel rilmenidine-derived compounds with anticancer potential and devoid of α2-adrenoceptor effects by means of ligand- and structure-based drug design approaches. Starting from a large virtual library, eleven compounds were selected, synthesized and submitted to biological evaluation. The most active compound 5 exhibited a cytotoxic profile similar to that of rilmenidine, but without appreciable affinity to α2-adrenoceptors. In addition, compound 5 significantly enhanced the apoptotic response to doxorubicin, and may thus represent an important tool for the development of better adjuvant chemotherapeutic strategies for doxorubicin-insensitive cancers.


Subject(s)
Adrenergic alpha-Agonists/pharmacology , Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Oxazoles/pharmacology , Apoptosis/drug effects , Drug Synergism , Humans , K562 Cells , Ligands , Molecular Structure , Receptors, Adrenergic, alpha-2/drug effects , Rilmenidine
2.
J Med Chem ; 64(9): 5931-5955, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33890770

ABSTRACT

Transient receptor potential cation channel subfamily M member 5 (TRPM5) is a nonselective monovalent cation channel activated by intracellular Ca2+ increase. Within the gastrointestinal system, TRPM5 is expressed in the stoma, small intestine, and colon. In the search for a selective agonist of TRPM5 possessing in vivo gastrointestinal prokinetic activity, a high-throughput screening was performed and compound 1 was identified as a promising hit. Hit validation and hit to lead activities led to the discovery of a series of benzo[d]isothiazole derivatives. Among these, compounds 61 and 64 showed nanomolar activity and excellent selectivity (>100-fold) versus related cation channels. The in vivo drug metabolism and pharmacokinetic profile of compound 64 was found to be ideal for a compound acting locally at the intestinal level, with minimal absorption into systemic circulation. Compound 64 was tested in vivo in a mouse motility assay at 100 mg/kg, and demonstrated increased prokinetic activity.


Subject(s)
Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Gastrointestinal Tract/metabolism , High-Throughput Screening Assays , TRPM Cation Channels/agonists , Animals , Benzothiazoles/metabolism , Benzothiazoles/pharmacokinetics , Drug Design , Gastrointestinal Tract/drug effects , Humans , Male , Mice , Molecular Targeted Therapy , Reproducibility of Results , Tissue Distribution
3.
J Med Chem ; 62(23): 10833-10847, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31729878

ABSTRACT

Cystic fibrosis (CF) is a multiorgan disease caused by mutations of the cystic fibrosis transmembrane conductance regulator (CFTR). In addition to respiratory impairment due to mucus accumulation, viruses and bacteria trigger acute pulmonary exacerbations, accelerating disease progression and mortality rate. Treatment complexity increases with patients' age, and simplifying the therapeutic regimen represents one of the key priorities in CF. We have recently reported the discovery of multitarget compounds able to "kill two birds with one stone" by targeting F508del-CFTR and PI4KIIIß and thus acting simultaneously as CFTR correctors and broad-spectrum enterovirus (EV) inhibitors. Starting from these preliminary results, we report herein a hit-to-lead optimization and multidimensional structure-activity relationship (SAR) study that led to compound 23a. This compound showed good antiviral and F508del-CFTR correction potency, additivity/synergy with lumacaftor, and a promising in vitro absorption, distribution, metabolism, and excretion (ADME) profile. It was well tolerated in vivo with no sign of acute toxicity and histological alterations in key biodistribution organs.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/drug therapy , Microsomes, Liver/drug effects , Animals , Antiviral Agents , Cell Survival/drug effects , Drug Delivery Systems , Humans , Male , Membranes, Artificial , Mice , Mice, Inbred C57BL , Permeability , Protein Binding , Serum Albumin, Human/chemistry , Serum Albumin, Human/metabolism , Toxicity Tests
4.
ChemMedChem ; 12(16): 1279-1285, 2017 08 22.
Article in English | MEDLINE | ID: mdl-28520140

ABSTRACT

Together with estrogen receptors ERα and ERß, the G protein-coupled estrogen receptor (GPER) mediates important pathophysiological signaling pathways induced by estrogens and is currently regarded as a promising target for ER-negative (ER-) and triple-negative (TN) breast cancer. Only a few selective GPER modulators have been reported to date, and their use in cancer cell lines has often led to contradictory results. Herein we report the application of virtual screening and cell-based studies for the identification of new chemical scaffolds with a specific antiproliferative effect against GPER-expressing breast cancer cell lines. Out of the four different scaffolds identified, 8-chloro-4-(4-chlorophenyl)pyrrolo[1,2-a]quinoxaline 14 c was found to be the most promising compound able to induce: 1) antiproliferative activity in GPER-expressing cell lines (MCF7 and SKBR3), similarly to G15; 2) no effect on cells that do not express GPER (HEK293); 3) a decrease in cyclin D1 expression; and 4) a sustained induction of cell-cycle negative regulators p53 and p21.


Subject(s)
Antineoplastic Agents/metabolism , Quinoxalines/metabolism , Receptors, G-Protein-Coupled/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/metabolism , Female , HEK293 Cells , Humans , MCF-7 Cells , Molecular Docking Simulation , Protein Structure, Tertiary , Quinoxalines/chemistry , Quinoxalines/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
5.
J Med Chem ; 60(4): 1400-1416, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28122178

ABSTRACT

Enteroviruses (EVs) are among the most frequent infectious agents in humans worldwide and represent the leading cause of upper respiratory tract infections. No drugs for the treatment of EV infections are currently available. Recent studies have also linked EV infection with pulmonary exacerbations, especially in cystic fibrosis (CF) patients, and the importance of this link is probably underestimated. The aim of this work was to develop a new class of multitarget agents active both as broad-spectrum antivirals and as correctors of the F508del-cystic fibrosis transmembrane conductance regulator (CFTR) folding defect responsible for >90% of CF cases. We report herein the discovery of the first small molecules able to simultaneously act as correctors of the F508del-CFTR folding defect and as broad-spectrum antivirals against a panel of EVs representative of all major species.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/drug therapy , Cystic Fibrosis/virology , Enterovirus/drug effects , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Drug Discovery , Enterovirus Infections/drug therapy , Enterovirus Infections/genetics , Enterovirus Infections/virology , Humans , Models, Molecular , Molecular Docking Simulation , Mutation , Protein Folding/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
6.
Expert Opin Drug Discov ; 10(6): 671-84, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25927601

ABSTRACT

INTRODUCTION: Maraviroc is a first-in-class antiretroviral (ARV) drug acting on a host cell target (CCR5), which blocks the entry of the HIV virus into the cell. Maraviroc is currently indicated for combination ARV treatment in adults infected only with CCR5-tropic HIV-1. AREAS COVERED: This drug discovery case history focuses on the key studies that led to the discovery and approval of maraviroc, as well as on post-launch clinical reports. The article is based on the data reported in published preclinical and clinical studies, conference posters and on drug package data. EXPERT OPINION: The profound understanding of HIV's entry mechanisms has provided a strong biological rationale for targeting the chemokine receptor CCR5. The CCR5-antagonist mariviroc, with its unique mode of action and excellent safety profile, is an important therapeutic option for HIV patients. In general, the authors believe that targeting host factors is a useful approach for combating new and re-emerging transmissible diseases, as well as pathogens that easily become resistant to common antiviral drugs. Maraviroc, offering a potent and safe cellular receptor-mediated pharmacological response to HIV, has paved the way for the development of a new generation of host-targeting antivirals.


Subject(s)
CCR5 Receptor Antagonists/therapeutic use , Cyclohexanes/therapeutic use , HIV Infections/drug therapy , Triazoles/therapeutic use , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , CCR5 Receptor Antagonists/administration & dosage , CCR5 Receptor Antagonists/pharmacology , Cyclohexanes/administration & dosage , Cyclohexanes/pharmacology , Drug Approval , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Drug Therapy, Combination , HIV Infections/virology , HIV-1/drug effects , HIV-1/isolation & purification , Humans , Maraviroc , Triazoles/administration & dosage , Triazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL