Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 365
1.
PLoS One ; 19(4): e0299198, 2024.
Article En | MEDLINE | ID: mdl-38635661

Herpesviruses have two distinct life cycle stages, latency and lytic replication. Epstein-Barr virus (EBV), a gamma-herpesvirus, establishes latency in vivo and in cultured cells. Cell lines harboring latent EBV can be induced into the lytic cycle by treatment with chemical inducing agents. In the Burkitt lymphoma cell line HH514-16 the viral lytic cycle is triggered by butyrate, a histone deacetylase (HDAC) inhibitor. Butyrate also alters expression of thousands of cellular genes. However, valproic acid (VPA), another HDAC inhibitor with global effects on cellular gene expression blocks EBV lytic gene expression in Burkitt lymphoma cell lines. Valpromide (VPM), an amide derivative of VPA, is not an HDAC inhibitor, but like VPA blocks induction of the EBV lytic cycle. VPA and VPM are the first examples of inhibitors of initial stages of lytic reactivation. We compared the effects of VPA and VPM, alone and in combination with butyrate, on host cellular gene expression using whole transcriptome analysis (RNA-seq). Gene expression was analyzed 6 h after addition of the compounds, a time before the first EBV lytic transcripts are detected. The results address two alternative, yet possibly complementary, mechanisms for regulation of EBV lytic reactivation. First, cellular genes that were up- or down-regulated by butyrate, but no longer altered in the presence of VPA or VPM, represent genes that correlated with EBV lytic reactivation. Second, genes regulated similarly by VPA and VPM in the absence and presence of butyrate are candidates for suppressors of EBV reactivation. Two genes upregulated by the lytic cycle inhibitors, CHAC1 and SLC7A11, are related to redox status and the iron-dependent cell death pathway ferroptosis. This study generates new hypotheses for control of the latency to lytic cycle switch of EBV and provides the first description of effects of the anti-convulsant drug VPM on global human cellular gene expression.


Burkitt Lymphoma , Epstein-Barr Virus Infections , Valproic Acid/analogs & derivatives , Humans , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Herpesvirus 4, Human/physiology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/metabolism , Epstein-Barr Virus Infections/drug therapy , Virus Activation , Gene Expression Profiling , Butyrates/pharmacology
2.
Viruses ; 13(12)2021 12 15.
Article En | MEDLINE | ID: mdl-34960791

Pseudorabies virus (PRV) infection of swine can produce Aujeszky's disease, which causes neurological, respiratory, and reproductive symptoms, leading to significant economic losses in the swine industry. Although humans are not the natural hosts of PRV, cases of human encephalitis and endophthalmitis caused by PRV infection have been reported between animals and workers. Currently, a lack of specific treatments and the emergence of new PRV strains against which existing vaccines do not protect makes the search for effective antiviral drugs essential. As an alternative to traditional nucleoside analogues such as acyclovir (ACV), we studied the antiviral effect of valpromide (VPD), a compound derived from valproic acid, against PRV infection in the PK15 swine cell line and the neuroblastoma cell line Neuro-2a. First, the cytotoxicity of ACV and VPD in cells was compared, demonstrating that neither compound was cytotoxic at a specific concentration range after 24 h exposure. Furthermore, the lack of direct virucidal effect of VPD outside of an infected cell environment was demonstrated. Finally, VPD was shown to have an antiviral effect on the viral production of two strains of pseudorabies virus (wild type NIA-3 and recombinant PRV-XGF) at the concentrations ranging from 0.5 to 1.5 mM, suggesting that VPD could be a suitable alternative to nucleoside analogues as an antiherpetic drug against Aujeszky's disease.


Antiviral Agents/pharmacology , Herpesvirus 1, Suid/drug effects , Pseudorabies/drug therapy , Valproic Acid/analogs & derivatives , Valproic Acid/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Mice , Neuroblastoma , Pseudorabies/virology , Swine , Swine Diseases/drug therapy , Swine Diseases/virology
3.
Article En | MEDLINE | ID: mdl-34419711

Laboratory measurements of intrinsic clearance support the development of TK models, with potential relevance to weight of evidence toxicity assessments of xenobiotics, including read-across, the concept of predictive estimation by data extrapolation between chemicals of similar structure (analogues). In this work a procedure with analytical method for determination of in vitro hepatic metabolic clearance, relevant to biotransformation toxicokinetic (TK) modelling, is presented. Cryopreserved primary human hepatocytes represent a suitable cells, due to their biological characteristics, for providing an in vitro model for simulating in vivo metabolic clearance. The experimental part considered an adequate sequential time-frame for collecting samples and controls for all chemicals tested, including centrifugation and aliquoting of the corresponding fractions until the instrumental session. For the first time, in vitro hepatocyte intrinsic clearance was measured for six analogue test chemicals: valproic acid, 2-ethyl caproic acid, octanoic acid, valeric acid, 2-methyl butyric acid and 2-trans pentenoic acid, during incubated cell culture exposure up to 2 h or 3.5 h. The time dependence of any metabolism was determined from analysis of the supernatant at intervals using a new developed analytical method for UPLC coupled with QTOF mass spectrometer. The chemicals could then be ranked by their relative intrinsic clearance. The analyses were reproducible, with coherence of the calculated in vitro intrinsic clearance between experiments.


Chromatography, High Pressure Liquid/methods , Hepatobiliary Elimination/physiology , Liver/metabolism , Mass Spectrometry/methods , Valproic Acid , Cells, Cultured , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Limit of Detection , Linear Models , Liver/cytology , Reproducibility of Results , Valproic Acid/analogs & derivatives , Valproic Acid/analysis , Valproic Acid/metabolism
4.
J Inorg Biochem ; 222: 111508, 2021 09.
Article En | MEDLINE | ID: mdl-34116426

Histone deacetylase inhibitors have often been used in combination treatment of various types of cancers due to their non-genotoxic epigenetic potential. Valproic acid (VPA) is a well-known histone deacetylase inhibitor. Conjugate of VPA with a phtoactive platinum diimine complex through an ester bond has been fabricated to potentiate the photocytotoxicity of the photosensitizer. Its capability to generate singlet oxygen, behavior in the presence of esterase, and photocytotoxicity in tumor cells have also been studied. The results revealed that the novel VPA-modified platinum diimine complex could produce singlet oxygen efficiently and release VPA in the presence of porcine liver esterase. The results also suggested that incorporation of VPA moiety into the platinum diimine complex might significantly enhance the cytotoxicity of the complex.


Coordination Complexes/pharmacology , Photosensitizing Agents/pharmacology , Valproic Acid/analogs & derivatives , Valproic Acid/pharmacology , Animals , Carboxylic Ester Hydrolases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/metabolism , Coordination Complexes/radiation effects , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/radiation effects , Humans , Light , Photochemotherapy , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/metabolism , Photosensitizing Agents/radiation effects , Platinum/chemistry , Prodrugs/chemical synthesis , Prodrugs/metabolism , Prodrugs/pharmacology , Prodrugs/radiation effects , Singlet Oxygen/metabolism , Swine , Valproic Acid/metabolism , Valproic Acid/radiation effects
5.
Eur J Pharmacol ; 903: 174150, 2021 Jul 15.
Article En | MEDLINE | ID: mdl-33961874

Inhibition of the activity of extracellular signal-regulated kinases (ERK1/2) induced by the activation of the dopamine D2 receptor signalling cascade may be a promising pharmacological target. The aim of this work was to study the involvement of ERK1/2 and dopamine D2 receptor in the mechanism of the anticonvulsant action of valproic acid (VA) and a new benzoylpyridine oxime derivative (GIZH-298), which showed antiepileptic activity in different models of epilepsy. We showed that subchronic exposure to maximal electroshock seizures (MES) for 5 days reduced the density of dopamine D2 receptors in the striatum of mice. GIZH-298 counteracted the decrease in the number of dopamine D2 receptors associated with MES and increased the number of ligand binding sites of dopamine D2 receptors in mice without MES. The affinity of dopamine D2 receptors to the ligand was not changed by GIZH-298. MES caused an increase in ERK1/2 and synapsin I phosphorylation in the striatum while GIZH-298, similar to VA, reduced the levels of both phospho-ERK1/2 and phosphosynapsin I after MES, which correlated with the decrease in the intensity of seizure in mice. In addition, GIZH-298 suppressed ERK1/2 phosphorylation in SH-SY5Y human neuroblastoma cells at therapeutic concentrations, while VA inhibited ERK1/2 phosphorylation in vivo but not in vitro. The data obtained expand the understanding of the mechanisms of action of VA and GIZH-298, which involve regulating the activity of ERK1/2 kinases, probably by modulating dopamine D2 receptors in limbic structures, as well as (in the case of GIZH-298) directly inhibiting of the ERK1/2 cascade.


Anticonvulsants/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/drug effects , Receptors, Dopamine D2/metabolism , Seizures/drug therapy , Valproic Acid/pharmacology , Animals , Anticonvulsants/administration & dosage , Cell Line, Tumor , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Electroshock/adverse effects , Humans , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neuroblastoma/metabolism , Phosphorylation/drug effects , Seizures/etiology , Seizures/metabolism , Synapsins/metabolism , Valproic Acid/administration & dosage , Valproic Acid/analogs & derivatives
6.
Int J Pharm ; 595: 120274, 2021 Feb 15.
Article En | MEDLINE | ID: mdl-33486026

The mechanical properties of powders determine the ease of manufacture and ultimately the quality of the oral solid dosage forms. Although poor mechanical properties of an active pharmaceutical ingredient (API) can be mitigated by using suitable excipients in a formulation, the effectiveness of that approach is limited for high dose drugs or multidrug tablets. In this context, improving the mechanical properties of the APIs through solid form optimisation is a good strategy to address such a challenge. This work explores the powder and tableting properties of various lamotrigine (LAM) solid forms with the aim to facilitate direct compression by overcoming the poor tabletability of LAM. The two drug-drug crystals of LAM with nicotinamide and valproic acid demonstrate superior flowability and tabletability over LAM. The improved powder properties are rationalised by structure analysis using energy framework, scanning electron microscopy, and Heckel analysis.


Drug Compounding/methods , Lamotrigine/chemistry , Powders/chemistry , Tablets/chemistry , Crystallography , Excipients/chemistry , Lamotrigine/analogs & derivatives , Microscopy, Electron, Scanning , Niacinamide/analogs & derivatives , Niacinamide/chemistry , Particle Size , Porosity , Pressure , Rheology , Spectrum Analysis, Raman , Valproic Acid/analogs & derivatives , Valproic Acid/chemistry , X-Ray Diffraction
7.
Molecules ; 27(1)2021 Dec 24.
Article En | MEDLINE | ID: mdl-35011339

Valproic acid (VPA) is a well-established anticonvulsant drug discovered serendipitously and marketed for the treatment of epilepsy, migraine, bipolar disorder and neuropathic pain. Apart from this, VPA has potential therapeutic applications in other central nervous system (CNS) disorders and in various cancer types. Since the discovery of its anticonvulsant activity, substantial efforts have been made to develop structural analogues and derivatives in an attempt to increase potency and decrease adverse side effects, the most significant being teratogenicity and hepatotoxicity. Most of these compounds have shown reduced toxicity with improved potency. The simple structure of VPA offers a great advantage to its modification. This review briefly discusses the pharmacology and molecular targets of VPA. The article then elaborates on the structural modifications in VPA including amide-derivatives, acid and cyclic analogues, urea derivatives and pro-drugs, and compares their pharmacological profile with that of the parent molecule. The current challenges for the clinical use of these derivatives are also discussed. The review is expected to provide necessary knowledgebase for the further development of VPA-derived compounds.


Molecular Structure , Valproic Acid/chemistry , Valproic Acid/pharmacology , Amides/chemistry , Amides/pharmacology , Animals , Anticonvulsants/administration & dosage , Anticonvulsants/chemistry , Anticonvulsants/pharmacology , Drug Monitoring , Epilepsy/drug therapy , Humans , Structure-Activity Relationship , Teratogens/chemistry , Teratogens/pharmacology , Urea/analogs & derivatives , Urea/chemistry , Urea/pharmacology , Valproic Acid/administration & dosage , Valproic Acid/analogs & derivatives
8.
Arch Toxicol ; 95(2): 641-657, 2021 02.
Article En | MEDLINE | ID: mdl-33111190

Since teratogenicity testing in mammals is a particular challenge from an animal welfare perspective, there is a great need for the development of alternative test systems. In this context, the zebrafish (Danio rerio) embryo has received increasing attention as a non-protected embryonic vertebrate in vivo model. The predictive power of zebrafish embryos for general vertebrate teratogenicity strongly depends on the correlation between fish and mammals with respect to both overall general toxicity and more specific endpoints indicative of certain modes-of-action. The present study was designed to analyze the correlation between (1) effects of valproic acid and nine of its analogues in zebrafish embryos and (2) their known neurodevelopmental effects in mice. To this end, zebrafish embryos exposed for 120 h in an extended version of the acute fish embryo toxicity test (FET; OECD TG 236) were analyzed with respect to an extended list of sublethal endpoints. Particular care was given to endpoints putatively related to neurodevelopmental toxicity, namely jitter/tremor, deformation of sensory organs (eyes) and craniofacial deformation, which might correlate to neural tube defects caused by valproic acid in mammals. A standard evaluation of lethal (LC according to OECD TG 236) and sublethal toxicity (EC) merely indicated that four out of ten compounds tested in zebrafish correlate with positive results in mouse in vivo studies. A detailed assessment of more specific effects, however, namely, jitter/tremor, small eyes and craniofacial deformation, resulted in a correspondence of 75% with in vivo mouse data. A refinement of endpoint analysis from an integration of all observations into one LCx or ECx data (as foreseen by current ecotoxicology-driven OECD guidelines) to a differential evaluation of endpoints specific of selected modes-of-action thus increases significantly the predictive power of the zebrafish embryo model for mammalian teratogenicity. However, for some of the endpoints observed, e.g., scoliosis, lordosis, pectoral fin deformation and lack of movement, further experiments are required for the identification of underlying modes-of-action and an unambiguous interpretation of their predictive power for mammalian toxicity.


Embryo, Nonmammalian/drug effects , Morphogenesis/drug effects , Valproic Acid/analogs & derivatives , Valproic Acid/toxicity , Zebrafish/embryology , Animals , Ecotoxicology , Lethal Dose 50 , Mice , Models, Biological , Neurodevelopmental Disorders/chemically induced , Neurotoxins/toxicity , Toxicity Tests, Acute
10.
Viruses ; 12(12)2020 11 26.
Article En | MEDLINE | ID: mdl-33256172

Herpes simplex viruses (HSVs) are neurotropic viruses with broad host range whose infections cause considerable health problems in both animals and humans. In fact, 67% of the global population under the age of 50 are infected with HSV-1 and 13% have clinically recurrent HSV-2 infections. The most prescribed antiherpetics are nucleoside analogues such as acyclovir, but the emergence of mutants resistant to these drugs and the lack of available vaccines against human HSVs has led to an imminent need for new antivirals. Valproic acid (VPA) is a branched short-chain fatty acid clinically used as a broad-spectrum antiepileptic drug in the treatment of neurological disorders, which has shown promising antiviral activity against some herpesviruses. Moreover, its amidic derivatives valpromide and valnoctamide also share this antiherpetic activity. This review summarizes the current research on the use of VPA and its amidic derivatives as alternatives to traditional antiherpetics in the fight against HSV infections.


Alphaherpesvirinae/drug effects , Amides/chemistry , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Valproic Acid/analogs & derivatives , Valproic Acid/pharmacology , Animals , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Molecular Structure
11.
J Alzheimers Dis ; 78(3): 907-910, 2020.
Article En | MEDLINE | ID: mdl-33074242

In a woman with Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) followed for 15 years, we observed magnetic resonance imaging white matter hyperintensities that vanished in the anterior temporal poles while the brain volume decreased unexpectedly. These imaging changes were transient and detected when the patient was being treated by valproic acid for stabilizing mood disturbances. This intriguing case supports that mechanisms underlying white matter hyperintensities can vary from one brain area to another and that important modifications of water influx into the brain tissue might be involved in some imaging features of CADASIL.


Antimanic Agents/therapeutic use , Bipolar Disorder/drug therapy , Brain/diagnostic imaging , CADASIL/diagnostic imaging , Leukoencephalopathies/diagnostic imaging , Lithium Compounds/therapeutic use , Valproic Acid/analogs & derivatives , Bipolar Disorder/complications , Brain/pathology , CADASIL/complications , Disease Progression , Drug Substitution , Female , Humans , Imaging, Three-Dimensional , Magnetic Resonance Imaging , Middle Aged , Organ Size , Temporal Lobe/diagnostic imaging , Temporal Lobe/pathology , Valproic Acid/therapeutic use
12.
J Pharm Sci ; 109(7): 2156-2165, 2020 07.
Article En | MEDLINE | ID: mdl-32240697

Different solid forms possess various physicochemical properties, which can significantly affect the stability, bioavailability, and manufacturability of the final product. DP-VPA, a complex of 1-stearoyl-2-valproyl-sn-glycero-3-phosphatidylcholine (DP-VPA-C18) and 1-palmitoyl-2-valproyl-sn-glycero-3-phosphatidylcholine (DP-VPA-C16), is currently under development as an antiepileptic drug. DP-VPA-C16 and DP-VPA-C18 crystallize together in solid solution forms. The solid forms of DP-VPA solid solution were studied herein. Powder X-ray diffraction (PXRD), thermogravimetric analysis (TGA), differential scanning calorimetry (DSC), attenuated total reflection Fourier transform infrared spectroscopy (ATR-FTIR), scanning electron microscopy (SEM), dynamic vapor sorption (DVS) and optical microscopy were used to characterize the different crystalline forms, known as polymorphs. The physicochemical properties, including hygroscopicity, thermodynamic behavior, and relative stability, of each form were investigated. DVS analysis showed that DP-VPA solid solution reduced the hygroscopicity of DP-VPA-C16. The relative humidity stability study revealed that Forms A and B are relatively stable, while Forms A-1, B-1, C and D are highly unstable under natural humidity. Further analysis revealed that Form A transforms into Form B through milling. Given the physicochemical properties of the available physical forms, Form B may be the optimal form for the formulation and development of antiepileptic drugs.


X-Ray Diffraction , Calorimetry, Differential Scanning , Microscopy, Electron, Scanning , Spectroscopy, Fourier Transform Infrared , Valproic Acid/analogs & derivatives
13.
Reprod Toxicol ; 93: 219-229, 2020 04.
Article En | MEDLINE | ID: mdl-32114065

In order to better explain, predict, or extrapolate to humans the developmental toxicity effects of chemicals to zebrafish (Danio rerio) embryos, we developed a physiologically-based pharmacokinetic (PBPK) model designed to predict organ concentrations of neutral or ionizable chemicals, up to 120 h post-fertilization. Chemicals' distribution is modeled in the cells, lysosomes, and mitochondria of ten organs of the embryo. The model's partition coefficients are calculated with sub-models using physicochemical properties of the chemicals of interest. The model accounts for organ growth and changes in metabolic clearance with time. We compared ab initio model predictions to data obtained on culture medium and embryo concentrations of valproic acid (VPA) and nine analogs during continuous dosing under the OECD test guideline 236. We further improved the predictions by estimating metabolic clearance and partition coefficients from the data by Bayesian calibration. We also assessed the performance of the model at reproducing data published by Brox et al. (2016) on VPA and 16 other chemicals. We finally compared dose-response relationships calculated for mortality and malformations on the basis of predicted whole embryo concentrations versus those based on nominal water concentrations. The use of target organ concentrations substantially shifted the magnitude of dose-response parameters and the relative toxicity ranking of chemicals studied.


Anticonvulsants/pharmacokinetics , Embryo, Nonmammalian/metabolism , Models, Biological , Valproic Acid/analogs & derivatives , Valproic Acid/pharmacokinetics , Zebrafish/metabolism , Animals
14.
Epilepsia ; 61(1): 149-156, 2020 01.
Article En | MEDLINE | ID: mdl-31957880

OBJECTIVES: To advance the development of (2S,3S)-sec-butylpropylacetamide (SPD) as a new treatment for acute repetitive seizures (ARS), by studying its pharmacokinetics (PK) in pigs and its PK-pharmacodynamic (PK-PD) correlation in rats. METHODS: Two (2S,3S)-SPD intramuscular formulations (FA and FB ) were administered to pigs and rats and blood samples were withdrawn at different times after dosing. Major PK parameters were estimated in both species. PD analysis was conducted in rats utilizing the maximal-electroshock seizure threshold (MEST) test. Because ARS treatment requires a rapid action, the MEST test allows comparative evaluation of (2S,3S)-SPD intramuscular injection on rat susceptibility to electroconvulsive shock at various times after dosing. RESULTS: In rats, (2S,3S)-SPD plasma exposure increased proportionally following intramuscular dosing (20, 25 and 40 mg/kg) of FA and FB . Peak plasma concentration (Cmax ) was obtained at 1-2 hours after dosing and ranged between 6.8 and 9.4 mg/L. (2S,3S)-SPD plasma concentration at 10 minutes after dosing (C10 ) ranged between 2.1 and 3.5 mg/mL, and its half-life ranged between 0.9 and 2.3 hours. The highest C10 value, which may indicate rapid activity onset, and the highest Cmax were observed following administration of FA (40 mg/kg): C10  = 3.5 mg/L and Cmax  = 9.5 mg/L. In the MEST test, (2S,3S)-SPD (20 and 60 mg/kg) significantly raised the tonic seizure threshold compared to vehicle at 4, 7, 10, and 20 minutes after dosing, with a 1.6-fold increase at 20 minutes, which coincided with (2S,3S)-SPD brain Cmax . Following intramuscular dosing of (2S,3S)-SPD (12 mg/kg) to pigs of FA and FB , a Cmax value of 0.9 mg/L was obtained 0.42 and 0.75 hours after dosing, respectively. (2S,3S)-SPD C10 was 0.27 mg/L (FA ) and 0.49 mg/L (FB ). (2S,3S)-SPD clearance, volume of distribution, and half-life were 2 L/h/kg, 18-28 L/kg, and 6.1-9.7 hours, respectively. SIGNIFICANCE: (2S,3S)-SPD demonstrated a good PK-PD correlation in the rat MEST test, with a rapid onset. (2S,3S)-SPD first PK study in pigs showed that doses >12 mg/kg are required to achieve in pigs the plasma concentrations associated with activity at the rat MEST test.


Amides/pharmacology , Anticonvulsants/pharmacology , Seizures , Valproic Acid/analogs & derivatives , Animals , Brain/drug effects , Female , Male , Rats , Rats, Sprague-Dawley , Stereoisomerism , Swine , Valproic Acid/pharmacology
15.
Carcinogenesis ; 41(7): 927-939, 2020 07 14.
Article En | MEDLINE | ID: mdl-31584613

New chemotherapeutic agents are needed for pancreatic cancer (PC). We have previously shown that phospho-valproic acid (MDC-1112) is effective in cell-line xenografts of PC. Here, we explored whether MDC-1112 is effective in additional clinically relevant animal models of PC and whether MDC-1112 enhances the anticancer effect of clinically used chemotherapeutic agents. MDC-1112 alone strongly reduced patient-derived pancreatic tumor xenograft growth, and extended survival of LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx1-Cre (KPC) mice. In both models, MDC-1112 inhibited STAT3 activation and its downstream signals, including Bcl-xL and cyclin D1. In human PC cell lines, P-V enhanced the growth inhibitory effect of gemcitabine (GEM), Abraxane and 5-FU, but not that of irinotecan. Normal human pancreatic epithelial cells were more resistant to the cytotoxic effects of MDC-1112/GEM combination. Furthermore, MDC-1112 enhanced GEM's effect on colony formation, apoptosis, cell migration, and cell invasion. In vivo, MDC-1112 and GEM, given alone, reduced patient-derived pancreatic tumor xenograft growth by 58% and 87%, respectively; whereas MDC-1112/GEM combination reduced tumor growth by 94%, inducing tumor stasis. In conclusion, MDC-1112 should be further explored as a potential agent to be used in combination with GEM for treating PC.


Abnormalities, Multiple/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Keratoconus/congenital , Organophosphates/pharmacology , Pancreatic Neoplasms/drug therapy , Valproic Acid/analogs & derivatives , Abnormalities, Multiple/pathology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Humans , Keratoconus/drug therapy , Keratoconus/pathology , Mice , Pancreatic Neoplasms/pathology , Signal Transduction/drug effects , Valproic Acid/pharmacology , Xenograft Model Antitumor Assays , Gemcitabine
16.
Reprod Toxicol ; 90: 44-52, 2019 12.
Article En | MEDLINE | ID: mdl-31445079

In vivo, neural crest (NC) cells contribute critically to heart formation. The embryonic stem cells in the cardiac Embryonic Stem cell Test (ESTc) differentiate into a heterogeneous cell population including non-cardiomyocyte cells. The use of molecular biomarkers from different mechanistic pathways can refine quantitative embryotoxicity assessment. Gene expression levels representing different signalling pathways that could relate to beating cardiomyocyte formation were analysed at different time-points. Immunocytochemistry showed NC cells were present in the ESTc and RT-qPCR showed upregulation of NC related gene expression levels in a time-dependent manner. NC related genes were sensitive to VPA and its analogues 2-ethylhexanoic acid (EHA) and 2-ethylhexanol (EHOL) and indicated VPA as the most potent one. STITCH ('search tool for interactions of chemicals') analysis showed relationships between the examined signalling pathways and suggested additional candidate marker genes. Biomarkers from dedicated mechanistic pathways, e.g. NC differentiation, provide promising tools for monitoring specific effects in ESTc.


Embryonic Stem Cells/drug effects , Gene Expression Regulation/drug effects , Myocytes, Cardiac/drug effects , Neural Crest/cytology , Valproic Acid/analogs & derivatives , Valproic Acid/toxicity , Animals , Cell Differentiation/drug effects , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Mice , Myocytes, Cardiac/metabolism , Tretinoin/metabolism
17.
Antiviral Res ; 168: 91-99, 2019 08.
Article En | MEDLINE | ID: mdl-31132386

Herpes simplex virus type 1 (HSV-1) is a ubiquitous infectious agent that can establish latency in neurons, and in some cases, viral retrograde transport results in infection of the central nervous system (CNS). Several antivirals have been identified with the ability to inhibit HSV-1 replication in human cells to a greater or lesser degree, most of which are nucleoside analogues that unfortunately exhibit teratogenic potential, embryotoxicity, carcinogenic or antiproliferative activities and resistances in immunocompromised patients, specially. In the present study, we assessed two amidic derivatives of valproic acid (VPA) - valpromide (VPD) and valnoctamide (VCD) - which are already used in clinic treatments, as feasible HSV-1 antivirals in glial cells. Both VPD and VCD have exhibited increased efficacy in bipolar disorders and as anticonvulsant drugs compared to VPA, while being less teratogenic and hepatotoxic. Cytotoxicity assays carried out in our laboratory showed that VPD and VCD were not toxic in a human oligodendroglioma cell line (HOG), at least at the concentrations established for human treatments. Infectivity assays showed a significant inhibition of HSV-1 infection in HOG cells after VPD and VCD treatment, being more pronounced in VPD-treated cells, comparable to the effects obtained with acyclovir. Furthermore, the same antiherpetic effects of VPD were observed in other oligodendrocytic cell lines and rat primary oligodendrocytes (OPCs), confirming the results obtained in HOG cells. Altogether, our results allow us to propose VPD as a potential antiherpetic drug that is able to act directly on oligodendrocytes of the CNS.


Amides/pharmacology , Antiviral Agents/pharmacology , Herpesvirus 1, Human/drug effects , Oligodendroglia/virology , Valproic Acid/analogs & derivatives , Amides/chemistry , Animals , Antiviral Agents/chemistry , Cell Survival/drug effects , Cells, Cultured , Humans , Molecular Structure , Oligodendroglia/drug effects , Rats , Valproic Acid/chemistry , Valproic Acid/pharmacology , Viral Proteins/genetics , Virus Internalization/drug effects , Virus Replication/drug effects
18.
Carcinogenesis ; 40(12): 1480-1491, 2019 12 31.
Article En | MEDLINE | ID: mdl-30994173

New therapeutic strategies against glioblastoma multiforme (GBM) are urgently needed. Signal transducer and activator of transcription 3 (STAT3), constitutively active in many GBM tumors, plays a major role in GBM tumor growth and represents a potential therapeutic target. We have documented previously that phospho-valproic acid (MDC-1112), which inhibits STAT3 activation, possesses strong anticancer properties in multiple cancer types. In this study, we explored the anticancer efficacy of MDC-1112 in preclinical models of GBM, and evaluated its mode of action. MDC-1112 inhibited the growth of multiple human GBM cell lines in a concentration- and time-dependent manner. Normal human astrocytes were resistant to MDC-1112, indicating selectivity. In vivo, MDC-1112 reduced the growth of subcutaneous GBM xenografts in mice by up to 78.2% (P < 0.01), compared with the controls. Moreover, MDC-1112 extended survival in an intracranial xenograft model. Although all vehicle-treated mice died by 19 days of treatment, 7 of 11 MDC-1112-treated mice were alive and healthy by the end of 5 weeks, with many showing tumor regression. Mechanistically, MDC-1112 inhibited STAT3 phosphorylation at the serine 727 residue, but not at tyrosine 705, in vitro and in vivo. STAT3 overexpression rescued GBM cells from the cell growth inhibition by MDC-1112. In addition, MDC-1112 reduced STAT3 levels in the mitochondria and enhanced mitochondrial levels of reactive oxygen species, which triggered apoptosis. In conclusion, MDC-1112 displays strong efficacy in preclinical models of GBM, with the serine 727 residue of STAT3 being its key molecular target. MDC-1112 merits further evaluation as a drug candidate for GBM. New therapeutic options are needed for glioblastoma. The novel agent MDC-1112 is an effective anticancer agent in multiple animal models of glioblastoma, and its mechanism of action involves the inhibition of STAT3 phosphorylation, primarily at its Serine 727 residue.


Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Glioblastoma/pathology , Organophosphates/pharmacology , STAT3 Transcription Factor/metabolism , Valproic Acid/analogs & derivatives , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Glioblastoma/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , STAT3 Transcription Factor/drug effects , Valproic Acid/pharmacology , Xenograft Model Antitumor Assays
19.
Drug Test Anal ; 11(7): 1035-1047, 2019 Jul.
Article En | MEDLINE | ID: mdl-30821115

DP-VPA is a phospholipid prodrug of valproic acid (VPA) that is developed as a potential treatment for epilepsy. To characterize the pharmacokinetics and excretion of DP-VPA, four reliable ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) methods were validated for quantitation of DP-VPA and its metabolite, VPA, in human plasma, urine, and feces. Protein precipitation and solid-phase extraction (SPE) were used for extraction of C16, C18 homologs of DP-VPA and VPA, respectively, from plasma. Urine and fecal homogenate involving the three analytes were efficiently prepared by methanol precipitation. The determinations of C16 DP-VPA, C18 DP-VPA, and VPA were performed using the positive multiple reaction monitoring (MRM) mode and the negative single ion monitoring (SIM) mode, respectively. The analytes were separated using gradient elution on C8 or phenyl column. Satisfactory results pertaining to selectivity, linearity, matrix effect, accuracy and precision, recovery, stability, dilution integrity, carryover, and incurred sample analysis (ISR) were obtained. The calibration ranges in human plasma were as follows: 0.00200-1.00 µg/mL for C16 DP-VPA, 0.0100-5.00 µg/mL for C18 DP-VPA, and 0.0500-20.0 µg/mL for VPA. The linear ranges in urine and fecal homogenate were 0.00500-2.00 µg/mL and 0.00200-0.800 µg/mL for C16 DP-VPA, 0.00500-2.00 µg/mL and 0.0100-4.00 µg/mL for C18 DP-VPA, and 0.200-80.0 µg/mL for VPA, respectively. The intra- and inter-batch coefficients of variation in three matrices ranged from 1.7% to 12.4% while the accuracy values ranged from 85.4% to 111.7%. The developed methods were successfully applied to determine pharmacokinetics of DP-VPA tablet after a single oral dose of 1200 mg in 12 healthy Chinese subjects under fed condition.


Anticonvulsants/pharmacokinetics , Valproic Acid/analogs & derivatives , Valproic Acid/pharmacokinetics , Adult , Anticonvulsants/blood , Anticonvulsants/urine , Chromatography, High Pressure Liquid/methods , Drug Monitoring/methods , Feces/chemistry , Female , Humans , Limit of Detection , Male , Tandem Mass Spectrometry/methods , Valproic Acid/blood , Valproic Acid/urine , Young Adult
20.
Epilepsia ; 60(2): 315-321, 2019 02.
Article En | MEDLINE | ID: mdl-30615805

OBJECTIVES: Children and adults are likely to be among the casualties in a civilian nerve agent exposure. This study evaluated the efficacy of valnoctamide (racemic-VCD), sec-butylpropylacetamide (racemic-SPD), and phenobarbital for stopping nerve agent seizures in both immature and adult rats. METHODS: Female and male postnatal day (PND) 21, 28, and 70 (adult) rats, previously implanted with electroencephalography (EEG) electrodes were exposed to seizure-inducing doses of the nerve agents sarin or VX and EEG was recorded continuously. Five minutes after seizure onset, animals were treated with SPD, VCD, or phenobarbital. The up-down method was used over successive animals to determine the anticonvulsant median effective dose (ED50 ) of the drugs. RESULTS: SPD-ED50 values in the VX model were the following: PND21, 53 mg/kg (male) and 48 mg/kg (female); PND28, 108 mg/kg (male) and 43 mg/kg (female); and PND70, 101 mg/kg (male) and 40 mg/kg (female). SPD-ED50 values in the sarin model were the following: PND21, 44 mg/kg (male) and 28 mg/kg (female); PND28, 79 mg/kg (male) and 34 mg/kg (female); and PND70, 53 mg/kg (male) and 53 mg/kg (female). VCD-ED50 values in the VX model were the following: PND21, 34 mg/kg (male) and 43 mg/kg (female); PND28, 165 mg/kg (male) and 59 mg/kg (female); and PND70, 87 mg/kg (male) and 91 mg/kg (female). VCD-ED50 values in the sarin model were the following: PND21, 45 mg/kg (male), 48 mg/kg (female); PND28, 152 mg/kg (male) 79 mg/kg (female); and PND70, 97 mg/kg (male) 79 mg/kg (female). Phenobarbital-ED50 values in the VX model were the following: PND21, 43 mg/kg (male) and 18 mg/kg (female); PND28, 48 mg/kg (male) and 97 mg/kg (female). Phenobarbital-ED50 values in the sarin model were the following: PND21, 32 mg/kg (male) and 32 mg/kg (female); PND28, 58 mg/kg (male) and 97 mg/kg (female); and PND70, 65 mg/kg (female). SIGNIFICANCE: SPD and VCD demonstrated anticonvulsant activity in both immature and adult rats in the sarin- and VX-induced status epilepticus models. Phenobarbital was effective in immature rats, whereas in adult rats, higher doses were required that were accompanied by toxicity. Overall, significantly less drug was required to stop seizures in PND21 animals than in the older animals, and overall, males required higher amounts of drug than females.


Amides/pharmacology , Seizures/drug therapy , Status Epilepticus/drug therapy , Valproic Acid/analogs & derivatives , Animals , Anticonvulsants/therapeutic use , Child , Disease Models, Animal , Electroencephalography/methods , Female , Humans , Male , Nerve Agents/pharmacology , Phenobarbital/therapeutic use , Rats , Valproic Acid/pharmacology
...