Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Colloid Interface Sci ; 627: 283-298, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35853406

ABSTRACT

HYPOTHESIS: The positive charge on liposome surface is known to promote the crossing of the Blood brain barrier (BBB). However, when diastereomeric cationic gemini amphiphiles are among lipid membrane components, also the stereochemistry may affect the permeability of the vesicle across the BBB. EXPERIMENTS: Liposomes featuring cationic diasteromeric gemini amphiphiles were formulated, characterized, and their interaction with cell culture models of BBB investigated. FINDINGS: Liposomes featuring the gemini amphiphiles were internalized in a monolayer of brain microvascular endothelial cells derived from human induced pluripotent stem cells (hiPSC) through an energy dependent transport, internalization involving both clathrin- and caveolae-mediated endocytosis. On the same formulations, the permeability was also evaluated across a human derived in vitro BBB transport model. The permeability of liposomes featuring the gemini amphiphiles was significantly higher compared to that of neutral liposomes (DPPC/Cholesterol), that were not able to cross BBB. Most importantly, the permeability was influenced by the stereochemistry of the gemini and pegylation of these formulations did not result in a drastic reduction of the crossing ability. The in vitro iPSC-derived BBB models used in this work represent an important advancement in the drug discovery research of novel brain delivery strategies and therapeutics for central nervous system diseases.


Subject(s)
Induced Pluripotent Stem Cells , Liposomes , Biological Transport , Blood-Brain Barrier , Cations , Cholesterol , Clathrin , Endothelial Cells , Humans , Liposomes/chemistry
2.
Int J Mol Sci ; 23(14)2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35887162

ABSTRACT

While blood-brain barrier (BBB) dysfunction has been described in neurological disorders, including Huntington's disease (HD), it is not known if endothelial cells themselves are functionally compromised when promoting BBB dysfunction. Furthermore, the underlying mechanisms of BBB dysfunction remain elusive given the limitations with mouse models and post mortem tissue to identify primary deficits. We established models of BBB and undertook a transcriptome and functional analysis of human induced pluripotent stem cell (iPSC)-derived brain-like microvascular endothelial cells (iBMEC) from HD patients or unaffected controls. We demonstrated that HD-iBMECs have abnormalities in barrier properties, as well as in specific BBB functions such as receptor-mediated transcytosis.


Subject(s)
Huntington Disease , Induced Pluripotent Stem Cells , Animals , Blood-Brain Barrier/physiology , Cell Differentiation , Endothelial Cells/physiology , Humans , Induced Pluripotent Stem Cells/physiology , Mice
3.
J Med Chem ; 63(24): 15785-15801, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33320012

ABSTRACT

Mutations in the human PANK2 gene are implicated in neurodegenerative diseases such as pantothenate kinase-associated neurodegeneration (PKAN) and result in low levels of coenzyme-A (CoA) in the CNS due to impaired production of phosphopantothenic acid (PPA) from vitamin B5. Restoration of central PPA levels by delivery of exogenous PPA is a recent strategy to reactivate CoA biosynthesis in PKAN patients. Fosmetpantotenate is an oral PPA prodrug. We report here the development of a new PANk2-/- knockout model that allows CoA regeneration in brain cells to be evaluated and describe two new series of cyclic phosphate prodrugs of PPA capable of regenerating excellent levels of CoA in this system. A proof-of-concept study in mouse demonstrates the potential of this new class of prodrugs to deliver PPA to the brain following oral administration and confirms incorporation of the prodrug-derived PPA into CoA.


Subject(s)
Pantothenic Acid/analogs & derivatives , Prodrugs/chemistry , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Coenzyme A/metabolism , Cyclization , Disease Models, Animal , Half-Life , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Lipid Droplets/chemistry , Lipid Droplets/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pantothenate Kinase-Associated Neurodegeneration/drug therapy , Pantothenate Kinase-Associated Neurodegeneration/pathology , Pantothenic Acid/chemistry , Pantothenic Acid/metabolism , Pantothenic Acid/therapeutic use , Prodrugs/metabolism , Prodrugs/therapeutic use , Structure-Activity Relationship
4.
Cells ; 9(4)2020 04 16.
Article in English | MEDLINE | ID: mdl-32316221

ABSTRACT

The blood-brain barrier (BBB) is responsible for the homeostasis between the cerebral vasculature and the brain and it has a key role in regulating the influx and efflux of substances, in healthy and diseased states. Stem cell technology offers the opportunity to use human brain-specific cells to establish in vitro BBB models. Here, we describe the establishment of a human BBB model in a two-dimensional monolayer culture, derived from human induced pluripotent stem cells (hiPSCs). This model was characterized by a transendothelial electrical resistance (TEER) higher than 2000 Ω∙cm2 and associated with negligible paracellular transport. The hiPSC-derived BBB model maintained the functionality of major endothelial transporter proteins and receptors. Some proprietary molecules from our central nervous system (CNS) programs were evaluated revealing comparable permeability in the human model and in the model from primary porcine brain endothelial cells (PBECs).


Subject(s)
Biological Transport/drug effects , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Animals , Astrocytes/metabolism , Biological Transport/physiology , Brain/cytology , Cell Differentiation/physiology , Cells, Cultured , Central Nervous System/chemistry , Central Nervous System/metabolism , Cryopreservation/methods , Humans , Immunohistochemistry , Permeability , Swine
5.
Mol Pharm ; 16(5): 2069-2082, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30916978

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disease caused by polyglutamine expansion in the huntingtin protein. For drug candidates targeting HD, the ability to cross the blood-brain barrier (BBB) and reach the site of action in the central nervous system (CNS) is crucial for achieving pharmacological activity. To assess the permeability of selected compounds across the BBB, we utilized a two-dimensional model composed of primary porcine brain endothelial cells and rat astrocytes. Our objective was to use this in vitro model to rank and prioritize compounds for in vivo pharmacokinetic and brain penetration studies. The model was first characterized using a set of validation markers chosen based on their functional importance at the BBB. It was shown to fulfill the major BBB characteristics, including functional tight junctions, high transendothelial electrical resistance, expression, and activity of influx and efflux transporters. The in vitro permeability of 54 structurally diverse known compounds was determined and shown to have a good correlation with the in situ brain perfusion data in rodents. We used this model to investigate the BBB permeability of a series of new HD compounds from different chemical classes, and we found a good correlation with in vivo brain permeation, demonstrating the usefulness of the in vitro model for optimizing CNS drug properties and for guiding the selection of lead compounds in a drug discovery setting.


Subject(s)
Blood-Brain Barrier/metabolism , Central Nervous System Agents/therapeutic use , Drug Discovery/methods , Huntington Disease/drug therapy , Models, Biological , ATP-Binding Cassette Transporters/metabolism , Animals , Astrocytes/metabolism , Capillary Permeability/physiology , Cells, Cultured , Cerebral Cortex/cytology , Coculture Techniques , Electric Impedance , Endothelial Cells/metabolism , Permeability , Rats , Rats, Sprague-Dawley , Solute Carrier Proteins/metabolism , Swine , Tight Junctions/metabolism
6.
J Pharmacol Exp Ther ; 341(2): 474-83, 2012 May.
Article in English | MEDLINE | ID: mdl-22338034

ABSTRACT

Anthracycline-related cardiotoxicity correlates with cardiac anthracycline accumulation and bioactivation to secondary alcohol metabolites or reactive oxygen species (ROS), such as superoxide anion (O2·â») and hydrogen peroxide H2O2). We reported that in an ex vivo human myocardial strip model, 3 or 10 µM amrubicin [(7S,9S)-9-acetyl-9-amino-7-[(2-deoxy-ß-D-erythro-pentopyranosyl)oxy]-7,8,9,10-tetrahydro-6,11-dihydroxy-5,12-napthacenedione hydrochloride] accumulated to a lower level compared with equimolar doxorubicin or epirubicin (J Pharmacol Exp Ther 341:464-473, 2012). We have characterized how amrubicin converted to ROS or secondary alcohol metabolite in comparison with doxorubicin (that formed both toxic species) or epirubicin (that lacked ROS formation and showed an impaired conversion to alcohol metabolite). Amrubicin and doxorubicin partitioned to mitochondria and caused similar elevations of H2O2, but the mechanisms of H2O2 formation were different. Amrubicin produced H2O2 by enzymatic reduction-oxidation of its quinone moiety, whereas doxorubicin acted by inducing mitochondrial uncoupling. Moreover, mitochondrial aconitase assays showed that 3 µM amrubicin caused an O2·â»-dependent reversible inactivation, whereas doxorubicin always caused an irreversible inactivation. Low concentrations of amrubicin therefore proved similar to epirubicin in sparing mitochondrial aconitase from irreversible inactivation. The soluble fraction of human myocardial strips converted doxorubicin and epirubicin to secondary alcohol metabolites that irreversibly inactivated cytoplasmic aconitase; in contrast, strips exposed to amrubicin failed to generate its secondary alcohol metabolite, amrubicinol, and only occasionally exhibited an irreversible inactivation of cytoplasmic aconitase. This was caused by competing pathways that favored formation and complete or near-to-complete elimination of 9-deaminoamrubicinol. These results characterize amrubicin metabolic advantages over doxorubicin and epirubicin, which may correlate with amrubicin cardiac safety in preclinical or clinical settings.


Subject(s)
Anthracyclines/metabolism , Anthracyclines/pharmacokinetics , Doxorubicin/metabolism , Doxorubicin/pharmacokinetics , Epirubicin/metabolism , Epirubicin/pharmacokinetics , Myocardium/metabolism , Aconitate Hydratase/metabolism , Alcohols/metabolism , Anthracyclines/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cytoplasm/metabolism , Doxorubicin/pharmacology , Epirubicin/pharmacology , Humans , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Troponin I/metabolism
7.
Eur J Med Chem ; 44(9): 3765-70, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19493593

ABSTRACT

The currently approved treatment for hepatitis C virus infections is a combination of Ribavirin and pegylated Interferon. It leads to a sustained virologic response in approximately only half of the patients treated. For this reason there is an urgent need of new therapeutic agents. 2'-C-Methylcytidine is the first nucleoside inhibitor of the HCV NS5B polymerase that was efficacious in reducing the viral load in patients infected with HCV. The application of a monophosphate prodrug approach based on unprecedented cyclic phosphoramidates is reported. Our SAR studies led to compounds that are efficiently converted to the active triphosphate in human hepatocytes.


Subject(s)
Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Cytidine/analogs & derivatives , Hepacivirus/drug effects , Hepatitis C/drug therapy , Prodrugs/pharmacology , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Cricetinae , Cytidine/administration & dosage , Cytidine/chemistry , Cytidine/metabolism , Cytidine/pharmacology , Drug Stability , Hepatocytes/virology , Humans , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/metabolism , Structure-Activity Relationship
8.
J Med Chem ; 52(11): 3453-6, 2009 Jun 11.
Article in English | MEDLINE | ID: mdl-19441846

ABSTRACT

The optimization of a potent, class I selective ketone HDAC inhibitor is shown. It possesses optimized pharmacokinetic properties in preclinical species, has a clean off-target profile, and is negative in a microbial mutagenicity (Ames) test. In a mouse xenograft model it shows efficacy comparable to that of vorinostat at a 10-fold reduced dose.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , Histone Deacetylase Inhibitors , Quinolines/pharmacokinetics , Animals , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Dogs , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , HeLa Cells , Humans , Mice , Quinolines/chemical synthesis , Rats
9.
J Med Chem ; 51(18): 5843-55, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18763751

ABSTRACT

Human immunodeficiency virus type-1 (HIV-1) integrase is one of the three virally encoded enzymes required for replication and therefore a rational target for chemotherapeutic intervention in the treatment of HIV-1 infection. We report here the discovery of Raltegravir, the first HIV-integrase inhibitor approved by FDA for the treatment of HIV infection. It derives from the evolution of 5,6-dihydroxypyrimidine-4-carboxamides and N-methyl-4-hydroxypyrimidinone-carboxamides, which exhibited potent inhibition of the HIV-integrase catalyzed strand transfer process. Structural modifications on these molecules were made in order to maximize potency as HIV-integrase inhibitors against the wild type virus, a selection of mutants, and optimize the selectivity, pharmacokinetic, and metabolic profiles in preclinical species. The good profile of Raltegravir has enabled its progression toward the end of phase III clinical trials for the treatment of HIV-1 infection and culminated with the FDA approval as the first HIV-integrase inhibitor for the treatment of HIV-1 infection.


Subject(s)
HIV Infections/drug therapy , HIV Integrase Inhibitors/pharmacology , Pyrrolidinones/pharmacology , Administration, Oral , Area Under Curve , Biological Availability , HIV Integrase Inhibitors/administration & dosage , HIV Integrase Inhibitors/pharmacokinetics , HIV Integrase Inhibitors/therapeutic use , Half-Life , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Pyrrolidinones/administration & dosage , Pyrrolidinones/pharmacokinetics , Pyrrolidinones/therapeutic use , Raltegravir Potassium
10.
J Med Chem ; 51(4): 861-74, 2008 Feb 28.
Article in English | MEDLINE | ID: mdl-18217703

ABSTRACT

HIV integrase is one of the three enzymes encoded by HIV genome and is essential for viral replication, but integrase inhibitors as marketed drugs have just very recently started to emerge. In this study, we show the evolution from the N-methylpyrimidinone structure to bicyclic pyrimidinones. Introduction of a suitably substituted amino moiety modulated the physical-chemical properties of the molecules and conferred nanomolar activity in the inhibition of spread of HIV-1 infection in cell culture. An extensive SAR study led to sulfamide (R)- 22b, which inhibited the strand transfer with an IC50 of 7 nM and HIV infection in MT4 cells with a CIC95 of 44 nM, and ketoamide (S)- 28c that inhibited strand transfer with an IC50 of 12 nM and the HIV infection in MT4 cells with a CIC95 of 13 nM and exhibited a good pharmacokinetic profile when dosed orally to preclinical species.


Subject(s)
Aminopyridines/chemical synthesis , Azepines/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/chemical synthesis , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase/metabolism , Pyrimidinones/chemical synthesis , Administration, Oral , Aminopyridines/pharmacokinetics , Aminopyridines/pharmacology , Animals , Azepines/pharmacokinetics , Azepines/pharmacology , Biological Availability , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line , Dogs , HIV Integrase/genetics , HIV Integrase Inhibitors/pharmacokinetics , HIV Integrase Inhibitors/pharmacology , HIV-1/drug effects , Humans , Macaca mulatta , Microsomes, Liver/metabolism , Pyrimidinones/pharmacokinetics , Pyrimidinones/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship
11.
J Med Chem ; 50(9): 2225-39, 2007 May 03.
Article in English | MEDLINE | ID: mdl-17428043

ABSTRACT

Human immunodeficiency virus type-1 (HIV-1) integrase, one of the three constitutive viral enzymes required for replication, is a rational target for chemotherapeutic intervention in the treatment of AIDS that has also recently been confirmed in the clinical setting. We report here on the design and synthesis of N-benzyl-5,6-dihydroxypyrimidine-4-carboxamides as a class of agents which exhibits potent inhibition of the HIV-integrase-catalyzed strand transfer process. In the current study, structural modifications on these molecules were made in order to examine effects on HIV-integrase inhibitory potencies. One of the most interesting compounds for this series is 2-[1-(dimethylamino)-1-methylethyl]-N-(4-fluorobenzyl)-5,6-dihydroxypyrimidine-4-carboxamide 38, with a CIC95 of 78 nM in the cell-based assay in the presence of serum proteins. The compound has favorable pharmacokinetic properties in preclinical species (rats, dogs, and monkeys) and shows no liabilities in several counterscreening assays, highlighting its potential as a clinically useful antiviral agent.


Subject(s)
HIV Integrase Inhibitors/chemical synthesis , HIV-1/drug effects , Pyridines/chemical synthesis , Pyrimidines/chemical synthesis , Animals , Biological Availability , Blood Proteins/metabolism , Cell Line, Tumor , Dogs , HIV Integrase Inhibitors/pharmacokinetics , HIV Integrase Inhibitors/pharmacology , Half-Life , Humans , Macaca mulatta , Protein Binding , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Structure-Activity Relationship , Virus Replication
12.
J Med Chem ; 49(18): 5404-7, 2006 Sep 07.
Article in English | MEDLINE | ID: mdl-16942012

ABSTRACT

A strategy to obtain a fully orthogonal estrogen-receptor-based gene switch responsive to molecules with acceptable pharmacological properties is presented. From a series of tetrahydrofluorenones active on the wild-type estrogen receptor (ER) an inactive analogue is chosen as a new lead compound. Coevolution of receptor mutants and ligands leads to an ER-based gene switch suitable for studies in animal models.


Subject(s)
Fluorenes/chemical synthesis , Receptors, Estrogen/drug effects , Binding Sites , Estradiol/chemistry , Estrogen Receptor alpha/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/drug effects , Estrogen Receptor beta/genetics , Fluorenes/chemistry , Fluorenes/pharmacology , HeLa Cells , Humans , Ligands , Models, Molecular , Molecular Structure , Mutation , Receptors, Estrogen/genetics , Structure-Activity Relationship
14.
Mol Ther ; 7(2): 271-80, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12597916

ABSTRACT

The tetracycline (Tc)-dependent system in its "on" version (rtTA system) displays a baseline activity in the uninduced state, severely limiting its potential applicability in human gene therapy. So far, two different strategies to circumvent this limitation have been described. On one side, co-expression of the tetracycline regulated repressor tTS(kid) has proved capable of substantially reducing the baseline activity of rtTA. On the other, novel versions of the activator, namely rtTA2(s)-S2 and rtTA2(s)-M2, with a lower basal activity have been engineered. We have combined these two approaches by co-expressing TS(kid) with the novel transactivators. Bicistronic vectors were constructed that co-express TS(kid) with rtTA, rtTA2(s)-S2, or rtTA2(s) M2, through an internal ribosome entry site (plasmids IRES-A, IRES-S2, and IRES-M2, respectively). IRES-M2 proved to be the most effective construct EX VIVO: it displayed a negligible basal activity, > 1000 fold inducibility, and high responsiveness to doxycycline (Dox). Upon delivery as plasmid DNA in mouse muscles, IRES-M2 facilitated 1000-fold induction of serum alkaline phosphatase (SEAP) gene expression and long-term, stringent, and strictly Dox-dose-dependent regulation of erythropoietin (Epo) gene expression. Tight regulation of the gene encoding SEAP was demonstrated also in non-human primates. Notably, the system was induced in animals by Dox-dosing regimens comparable to those used in humans.


Subject(s)
Doxycycline/pharmacology , Genetic Vectors , Transcription, Genetic , Animals , Anti-Bacterial Agents/pharmacology , Gene Expression Regulation , HeLa Cells , Humans , Macaca mulatta , Mice , Mice, Inbred BALB C , Plasmids/metabolism , Tetracycline/pharmacology , Time Factors , Transcriptional Activation , Transfection , Transgenes , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...