Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
2.
Biomolecules ; 13(9)2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37759709

ABSTRACT

While tumor-associated macrophages (TAM) have pro-tumoral activity, the ablation of macrophages in cancer may be undesirable since they also have anti-tumoral functions, including T cell priming and activation against tumor antigens. Alendronate is a potent amino-bisphosphonate that modulates the function of macrophages in vitro, with potential as an immunotherapy if its low systemic bioavailability can be addressed. We repurposed alendronate in a non-leaky and long-circulating liposomal carrier similar to that of the clinically approved pegylated liposomal doxorubicin to facilitate rapid clinical translation. Here, we tested liposomal alendronate (PLA) as an immunotherapeutic agent for cancer in comparison with a standard of care immunotherapy, a PD-1 immune checkpoint inhibitor. We showed that the PLA induced bone marrow-derived murine non-activated macrophages and M2-macrophages to polarize towards an M1-functionality, as evidenced by gene expression, cytokine secretion, and lipidomic profiles. Free alendronate had negligible effects, indicating that liposome encapsulation is necessary for the modulation of macrophage activity. In vivo, the PLA showed significant accumulation in tumor and tumor-draining lymph nodes, sites of tumor immunosuppression that are targets of immunotherapy. The PLA remodeled the tumor microenvironment towards a less immunosuppressive milieu, as indicated by a decrease in TAM and helper T cells, and inhibited the growth of established tumors in the B16-OVA melanoma model. The improved bioavailability and the beneficial effects of PLA on macrophages suggest its potential application as immunotherapy that could synergize with T-cell-targeted therapies and chemotherapies to induce immunogenic cell death. PLA warrants further clinical development, and these clinical trials should incorporate tumor and blood biomarkers or immunophenotyping studies to verify the anti-immunosuppressive effect of PLA in humans.

3.
Sci Rep ; 13(1): 8951, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37268656

ABSTRACT

Fuel influx and metabolism replenish carbon lost during normal neural activity. Ketogenic diets studied in epilepsy, dementia and other disorders do not sustain such replenishment because their ketone body derivatives contain four carbon atoms and are thus devoid of this anaplerotic or net carbon donor capacity. Yet, in these diseases carbon depletion is often inferred from cerebral fluorodeoxyglucose-positron emission tomography. Further, ketogenic diets may prove incompletely therapeutic. These deficiencies provide the motivation for complementation with anaplerotic fuel. However, there are few anaplerotic precursors consumable in clinically sufficient quantities besides those that supply glucose. Five-carbon ketones, stemming from metabolism of the food supplement triheptanoin, are anaplerotic. Triheptanoin can favorably affect Glucose transporter type 1 deficiency (G1D), a carbon-deficiency encephalopathy. However, the triheptanoin constituent heptanoate can compete with ketogenic diet-derived octanoate for metabolism in animals. It can also fuel neoglucogenesis, thus preempting ketosis. These uncertainties can be further accentuated by individual variability in ketogenesis. Therefore, human investigation is essential. Consequently, we examined the compatibility of triheptanoin at maximum tolerable dose with the ketogenic diet in 10 G1D individuals using clinical and electroencephalographic analyses, glycemia, and four- and five-carbon ketosis. 4 of 8 of subjects with pre-triheptanoin beta-hydroxybutyrate levels greater than 2 mM demonstrated a significant reduction in ketosis after triheptanoin. Changes in this and the other measures allowed us to deem the two treatments compatible in the same number of individuals, or 50% of persons in significant beta-hydroxybutyrate ketosis. These results inform the development of individualized anaplerotic modifications to the ketogenic diet.ClinicalTrials.gov registration NCT03301532, first registration: 04/10/2017.


Subject(s)
Diet, Ketogenic , Ketosis , Animals , Humans , Glucose Transporter Type 1 , 3-Hydroxybutyric Acid , Ketone Bodies
4.
Sci Rep ; 13(1): 3465, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36859467

ABSTRACT

Augmentation of anaplerosis, or replenishment of carbon lost during intermediary metabolic transitions, is desirable in energy metabolism defects. Triheptanoin, the triglyceride of 7-carbon heptanoic acid, is anaplerotic via direct oxidation or 5-carbon ketone body generation. In this context, triheptanoin can be used to treat Glucose transporter type 1 deficiency encephalopathy (G1D). An oral triheptanoin dose of 1 g/Kg/day supplies near 35% of the total caloric intake and impacted epilepsy and cognition in G1D. This provided the motivation to establish a maximum, potentially greater dose. Using a 3 + 3 dose-finding approach useful in oncology, we studied three age groups: 4-6, 6.8-10 and 11-16 years old. This allowed us to arrive at a maximum tolerated dose of 45% of daily caloric intake for each group. Safety was ascertained via analytical blood measures. One dose-limiting toxicity, occurring in 1 of 6 subjects, was encountered in the middle age group in the context of frequently reduced gastrointestinal tolerance for all groups. Ketonemia following triheptanoin was determined in another group of G1D subjects. In them, ß-ketopentanoate and ß-hydroxypentanoate concentrations were robustly but variably increased. These results enable the rigorous clinical investigation of triheptanoin in G1D by providing dosing and initial tolerability, safety and ketonemic potential.ClinicalTrials.gov registration: NCT03041363, first registration 02/02/2017.


Subject(s)
Ketosis , Middle Aged , Humans , Child, Preschool , Glucose Transporter Type 1 , Carbon , Triglycerides
6.
Sci Rep ; 12(1): 15503, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36109613

ABSTRACT

Gyriform mammals display neurophysiological and neural network activity that other species exhibit only in rudimentary or dissimilar form. However, neural recordings from large mammals such as the pig can be anatomically hindered and pharmacologically suppressed by anesthetics. This curtails comparative inferences. To mitigate these limitations, we set out to modify electrocorticography, intracerebral depth and intracortical recording methods to study the anesthetized pig. In the process, we found that common forms of infused anesthesia such as pentobarbital or midazolam can be neurophysiologic suppressants acting in dose-independent fashion relative to anesthetic dose or brain concentration. Further, we corroborated that standard laboratory conditions may impose electrical interference with specific neural signals. We thus aimed to safeguard neural network integrity and recording fidelity by developing surgical, anesthesia and noise reduction methods and by working inside a newly designed Faraday cage, and evaluated this from the point of view of neurophysiological power spectral density and coherence analyses. We also utilized novel silicon carbide electrodes to minimize mechanical disruption of single-neuron activity. These methods allowed for the preservation of native neurophysiological activity for several hours. Pig electrocorticography recordings were essentially indistinguishable from awake human recordings except for the small segment of electrical activity associated with vision in conscious persons. In addition, single-neuron and paired-pulse stimulation recordings were feasible simultaneously with electrocorticography and depth electrode recordings. The spontaneous and stimulus-elicited neuronal activities thus surveyed can be recorded with a degree of precision similar to that achievable in rodent or any other animal studies and prove as informative as unperturbed human electrocorticography.


Subject(s)
Anesthetics , Wakefulness , Animals , Brain/physiology , Humans , Mammals , Midazolam , Neurons/physiology , Pentobarbital , Swine
7.
J Pharm Biomed Anal ; 205: 114335, 2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34482182

ABSTRACT

Interest in human and experimental animal metabolism of substrates containing an odd number of carbons capable of fueling the tricarboxylic acid cycle such as heptanoic acid has motivated us to develop and validate a selective and specific liquid chromatographytandem mass spectrometric method for the simultaneous, quantitative determination of the ketone body byproducts 3-hydroxypentanoic acid and 3-oxopentanoic acid in plasma. Human plasma samples were protein-precipitated with methanol containing 0.2% formic acid. Chromatographic resolution was achieved on a Phenomenex Luna C18 column using gradient elution with mobile phases of water containing 0.1% formic acid and methanol containing 0.1% formic acid at 0.3 mL/min flow rate. The retention times of 3-hydroxypentanoic acid, 3-oxopentanoic acid and sulbactam (internal standard) were 3.85, 4.23, and 5.11 min, respectively. Validation was conducted in accordance with United States Food and Drug Administration guidance. The validated range of 3-hydroxypentanoic acid was 0.078-5 µg/mL and 0.156-10 µg/mL for 3-oxopentanoic acid. The method was accurate and precise over this range and exhibited 10-fold dilution integrity in human plasma. Recovery> 88% was achieved for analytes and internal standard. There was no matrix effect observed in human plasma. Both 3-hydroxypentanoic acid and 3-oxopentanoic acid were stable across conditions including autosampler, benchtop and freeze-thaw, as well as demonstrated long-term stability at -80 °C. The method was applied to the measurement of 3-hydroxypentanoic acid and 3-oxopentanoic acid concentrations in plasma from subjects receiving the triglyceride triheptanoin (as a source of heptanoate) for the experimental treatment of glucose transporter type I deficiency (G1D) syndrome.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/blood , Monosaccharide Transport Proteins/deficiency , Organic Chemicals , Tandem Mass Spectrometry , Animals , Chromatography, Liquid , Glucose Transport Proteins, Facilitative , Humans , Monosaccharide Transport Proteins/blood , Reproducibility of Results
8.
Mol Cancer Ther ; 20(10): 1904-1915, 2021 10.
Article in English | MEDLINE | ID: mdl-34376577

ABSTRACT

Itraconazole, an FDA-approved antifungal, has antitumor activity against a variety of cancers. We sought to determine the effects of itraconazole on esophageal cancer and elucidate its mechanism of action. Itraconazole inhibited cell proliferation and induced G1-phase cell-cycle arrest in esophageal squamous cell carcinoma and adenocarcinoma cell lines. Using an unbiased kinase array, we found that itraconazole downregulated protein kinase AKT phosphorylation in OE33 esophageal adenocarcinoma cells. Itraconazole also decreased phosphorylation of downstream ribosomal protein S6, transcriptional expression of the upstream receptor tyrosine kinase HER2, and phosphorylation of upstream PI3K in esophageal cancer cells. Lapatinib, a tyrosine kinase inhibitor that targets HER2, and siRNA-mediated knockdown of HER2 similarly suppressed cancer cell growth in vitro Itraconazole significantly inhibited growth of OE33-derived flank xenografts in mice with detectable levels of itraconazole and its primary metabolite, hydroxyitraconazole, in esophagi and tumors. HER2 total protein and phosphorylation of AKT and S6 proteins were decreased in xenografts from itraconazole-treated mice compared to xenografts from placebo-treated mice. In an early phase I clinical trial (NCT02749513) in patients with esophageal cancer, itraconazole decreased HER2 total protein expression and phosphorylation of AKT and S6 proteins in tumors. These data demonstrate that itraconazole has potent antitumor properties in esophageal cancer, partially through blockade of HER2/AKT signaling.


Subject(s)
Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Itraconazole/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Receptor, ErbB-2/antagonists & inhibitors , Animals , Apoptosis , Cell Cycle , Cell Movement , Cell Proliferation , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Female , Humans , Itraconazole/pharmacokinetics , Maximum Tolerated Dose , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , Tissue Distribution , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Eur J Med Chem ; 209: 112866, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33039722

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive type of cancer characterized by higher metastatic and reoccurrence rates, where approximately one-third of TNBC patients suffer from the metastasis in the brain. At the same time, TNBC shows good responses to chemotherapy, a feature that fuels the search for novel compounds with therapeutic potential in this area. Recently, we have identified novel urea-based compounds with cytotoxicity against selected cell lines and with the ability to cross the blood-brain barrier in vivo. We have synthesized and analyzed a library of more than 40 compounds to elucidate the key features responsible for the observed activity. We have also identified FGFR1 as a molecular target that is affected by the presence of these compounds, confirming our data using in silico model. Overall, we envision that these compounds can be further developed for the potential treatment of metastatic breast cancer.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Urea/analogs & derivatives , Urea/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Drug Design , Female , Humans , Mice, Inbred C57BL , Molecular Docking Simulation , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Structure-Activity Relationship , Triple Negative Breast Neoplasms/metabolism , Urea/pharmacokinetics
11.
Article in English | MEDLINE | ID: mdl-32905989

ABSTRACT

The purpose of this work was to develop and validate a single sensitive, selective and rapid bioanalytical method to determine ceftolozane and tazobactam concentrations in human plasma and urine and to use this method to analyze samples from a human clinical study. Human plasma and urine samples were prepared by protein precipitation using a solution of acetonitrile, water and formic acid. Following protein precipitation, samples were analyzed by liquid chromatography tandem mass spectrometry. Chromatographic resolution was achieved on a Kinetex PFP column using a gradient elution, a flow rate of 0.4 mL/min, and a total run time of 5 min. Positive electrospray ionization was employed and analytes were quantitated using multi-reaction monitoring mode. Method validation was conducted in accordance with Unites States Food and Drug Administration's regulatory guidelines for bioanalytical method validation. Calibration curves were determined to linear over the range of 0.1 to 40 µg/mL for ceftolozane and 0.05 to 20 µg/mL for tazobactam. The method was determined to be accurate (-6.24 to 12.53 percent relative error), precise (less than 13.28 percent standard deviation) and sensitive in both human plasma and urine. Ceftolozane and tazobactam were determined to be stable across a battery of stability studies including autosampler, benchtop, freeze/thaw and long-term stability. This validated method successfully applied to human clinical samples to determine the concentration versus time profiles of the intravenously administered combination of Zerbaxa (ceftolozane-tazobactam) in burn patients.


Subject(s)
Cephalosporins , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Tazobactam , Cephalosporins/administration & dosage , Cephalosporins/blood , Cephalosporins/pharmacokinetics , Cephalosporins/urine , Humans , Limit of Detection , Linear Models , Reproducibility of Results , Tazobactam/administration & dosage , Tazobactam/blood , Tazobactam/pharmacokinetics , Tazobactam/urine
12.
Clin Cancer Res ; 26(22): 6017-6027, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32847935

ABSTRACT

PURPOSE: Itraconazole has been repurposed as an anticancer therapeutic agent for multiple malignancies. In preclinical models, itraconazole has antiangiogenic properties and inhibits Hedgehog pathway activity. We performed a window-of-opportunity trial to determine the biologic effects of itraconazole in human patients. EXPERIMENTAL DESIGN: Patients with non-small cell lung cancer (NSCLC) who had planned for surgical resection were administered with itraconazole 300 mg orally twice daily for 10-14 days. Patients underwent dynamic contrast-enhanced MRI and plasma collection for pharmacokinetic and pharmacodynamic analyses. Tissues from pretreatment biopsy, surgical resection, and skin biopsies were analyzed for itraconazole and hydroxyitraconazole concentration, and vascular and Hedgehog pathway biomarkers. RESULTS: Thirteen patients were enrolled in this study. Itraconazole was well-tolerated. Steady-state plasma concentrations of itraconazole and hydroxyitraconazole demonstrated a 6-fold difference across patients. Tumor itraconazole concentrations trended with and exceeded those of plasma. Greater itraconazole levels were significantly and meaningfully associated with reduction in tumor volume (Spearman correlation, -0.71; P = 0.05) and tumor perfusion (Ktrans; Spearman correlation, -0.71; P = 0.01), decrease in the proangiogenic cytokines IL1b (Spearman correlation, -0.73; P = 0.01) and GM-CSF (Spearman correlation, -1.00; P < 0.001), and reduction in tumor microvessel density (Spearman correlation, -0.69; P = 0.03). Itraconazole-treated tumors also demonstrated distinct metabolic profiles. Itraconazole treatment did not alter transcription of GLI1 and PTCH1 mRNA. Patient size, renal function, and hepatic function did not predict itraconazole concentrations. CONCLUSIONS: Itraconazole demonstrates concentration-dependent early antivascular, metabolic, and antitumor effects in patients with NSCLC. As the number of fixed dose cancer therapies increases, attention to interpatient pharmacokinetics and pharmacodynamics differences may be warranted.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Itraconazole/administration & dosage , Neovascularization, Pathologic/drug therapy , Adult , Angiogenesis Inhibitors/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Biopsy , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/surgery , Female , Hedgehog Proteins/genetics , Humans , Itraconazole/analogs & derivatives , Itraconazole/blood , Itraconazole/pharmacokinetics , Magnetic Resonance Imaging , Male , Middle Aged , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/surgery , Patched-1 Receptor/genetics , Zinc Finger Protein GLI1/genetics
13.
J Pharm Biomed Anal ; 188: 113466, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32668395

ABSTRACT

The purpose of this work was to develop and validate a rapid, sensitive and robust liquid chromatography tandem mass spectrometric method for the quantification of ß-lapachone in human plasma and to use that method to analyze human clinical samples. Sample preparation for the developed method involved liquid-liquid extraction using ethyl acetate for extraction of ß-lapachone and cryptotanshinone (internal standard) from human plasma. Chromatographic resolution was achieved on a Kinetex C18 column using a gradient elution and a chromatographic flow rate of 0.5 mL/min. The retention times of ß-lapachone and cryptotanshinone were 1.98 and 2.28 min, respectively, and the method had a total run time of 4 min. Bioanalytical method validation was conducted in accordance with the United States Food and Drug Administration regulatory guidelines. The method was validated over 2 calibration ranges in order to support high- and low-dose clinical studies. Calibration curve-1 covered the range of 0.25-50 ng/mL and calibration curve-2 covered the range of 50-2000 ng/mL. The method was determined to be accurate (percent relative errors between -1.07 to 5.36 %), precise (percent relative standard deviations less than 7.4), and sensitive (LLOQ 0.25 ng/mL). ß-lapachone was determined to be stable (% change from time = 0 between -11.6 and 12.6 %) across the autosampler, benchtop, freeze/thaw and long-term (63 days) stability studies. The validated bioanalytical method was employed to determine ß-lapachone concentrations in human plasma samples from a clinical study.


Subject(s)
Liquid-Liquid Extraction , Tandem Mass Spectrometry , Chromatography, High Pressure Liquid , Chromatography, Liquid , Humans , Naphthoquinones , Reproducibility of Results
14.
Breastfeed Med ; 14(2): 98-101, 2019 03.
Article in English | MEDLINE | ID: mdl-30431332

ABSTRACT

BACKGROUND: Oral mesalamine (5-amino salicylic acid [5-ASA]) is an anti-inflammatory agent commonly used to treat inflammatory bowel disease such as ulcerative colitis and Crohn's disease. The transfer of mesalamine into human milk has to date been poorly described at the current dosages and newer formulations. This study was designed to determine transfer of mesalamine into human milk as a function of maternal dose and time, and dosage form. STUDY DESIGN: Ten breastfeeding mothers (age 28-41 years) suffering from inflammatory bowel disease were recruited who provided milk samples at 0, 1, 2, 4, 8, 12, and 24 hours after a single daily dose of oral mesalamine in pH-dependent gastroresistant coated tablets (1.2, 2.4, 3.6, and 4.8 g). Milk samples were analyzed using liquid chromatography/tandem mass spectrometry. RESULTS: A total of 10 women were enrolled for the study. The calibration curve for mesalamine was linear over a concentration range of 0.32-200 ng/mL. Irrespective of maternal dose, mesalamine levels in milk were exceedingly low. However, a wide range of mesalamine levels were observed among all the participants. The relative infant doses were all lower than 0.1% (range 0.003-0.085%). CONCLUSION: Regardless of dose and high variability, mesalamine levels in human milk were present in exceedingly low levels. The mothers in this study reported no side effects with their infants. These results suggest that the transfer of mesalamine into milk is very low and poses minimal risks to the breastfed infant.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Breast Feeding , Inflammatory Bowel Diseases/drug therapy , Mesalamine/administration & dosage , Milk, Human/chemistry , Administration, Oral , Adult , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Female , Humans , Linear Models , Mesalamine/pharmacokinetics
15.
Br J Cancer ; 119(8): 928-936, 2018 10.
Article in English | MEDLINE | ID: mdl-30318513

ABSTRACT

BACKGROUND: NAD(P)H:quinone oxidoreductase 1 (NQO1) is a two-electron oxidoreductase expressed in multiple tumour types. ARQ 761 is a ß-lapachone (ß-lap) analogue that exploits the unique elevation of NQO1 found in solid tumours to cause tumour-specific cell death. METHODS: We performed a 3+3 dose escalation study of 3 schedules (weekly, every other week, 2/3 weeks) of ARQ 761 in patients with refractory advanced solid tumours. Tumour tissue was analysed for NQO1 expression. After 20 patients were analysed, enrolment was restricted to patients with NQO1-high tumours (H-score ≥ 200). RESULTS: A total of 42 patients were treated. Median number of prior lines of therapy was 4. Maximum tolerated dose was 390 mg/m2 as a 2-h infusion every other week. Dose-limiting toxicity was anaemia. The most common treatment-related adverse events were anaemia (79%), fatigue (45%), hypoxia (33%), nausea (17%), and vomiting (17%). Transient grade 3 hypoxia, reflecting possible methemoglobinaemia, occurred in 26% of patients. Among 32 evaluable patients, best response was stable disease (n = 12); 6 patients had tumour shrinkage. There was a trend towards improved efficacy in NQO1-high tumours (P = 0.06). CONCLUSIONS: ARQ 761 has modest single-agent activity, which appears associated with tumour NQO1 expression. Principal toxicities include anaemia and possible methemoglobinaemia.


Subject(s)
Apoptosis/drug effects , NAD(P)H Dehydrogenase (Quinone)/analysis , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Naphthoquinones/therapeutic use , Necrosis/chemically induced , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , DNA Damage/drug effects , Female , Humans , Male , Middle Aged , Naphthoquinones/chemistry , Reactive Oxygen Species/metabolism
16.
17.
Am J Physiol Endocrinol Metab ; 314(4): E322-E333, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29138223

ABSTRACT

Maternal obesity in pregnancy has been linked to a spectrum of adverse developmental changes. Involvement of eCBs in obesity is well characterized. However, information regarding eCB physiology in obesity associated with pregnancy is sparse. This study evaluated fetomaternal hepatic, systemic, and placental eCB molecular changes in response to maternal consumption of a HFD. From ≥9 mo before conception, nonpregnant baboons ( Papio spp.) were fed a diet of either 45 (HFD; n = 11) or 12% fat or a control diet (CTR; n = 11), and dietary intervention continued through pregnancy. Maternal and fetal venous plasma samples were evaluated using liquid chromatography-mass spectrometry to quantify AEA and 2-AG. Placental, maternal and fetal hepatic tissues were analyzed using RT-PCR, Western blot, and immunohistochemistry. mRNA and protein expression of endocannabinoid receptors (CB1R and CB2R), FAAH, DAGL, MAGL, and COX-2 were determined. Statistical analyses were performed with the nonparametric Scheirer-Ray-Hare extension of the Kruskal-Wallis test to analyze the effects of diet (HFD vs. CTR), fetal sex (male vs. female), and the diet × sex interaction. Fetal weight was influenced by fetal sex but not by maternal diet. The increase in maternal weight in animals fed the HFD vs. the CTR diet approached significance ( P = 0.055). Maternal circulating 2-AG concentrations increased, and fetal circulating concentrations decreased in the HFD group, independently of fetal sex. CB1R receptor expression was detected in syncytiotrophoblasts (HFD) and the fetal endothelium (CTR and HFD). Placental CB2R protein expression was higher in males and lower in female fetuses in the HFD group. Fetal hepatic CB2R, FAAH, COX-2 (for both fetal sexes), and DAGLα (in male fetuses) protein expression decreased in the HFD group compared with the CTR group. We conclude that consumption of a HFD during pregnancy results in fetal systemic 2-AG and hepatic eCB deficiency.


Subject(s)
Diet, High-Fat , Dietary Fats/pharmacology , Endocannabinoids/metabolism , Liver/drug effects , Maternal Nutritional Physiological Phenomena , Placenta/drug effects , Animals , Diet, High-Fat/adverse effects , Female , Liver/metabolism , Male , Maternal Nutritional Physiological Phenomena/drug effects , Maternal-Fetal Exchange/drug effects , Papio , Placenta/metabolism , Pregnancy , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/drug effects
18.
BMC Neurosci ; 18(1): 71, 2017 10 11.
Article in English | MEDLINE | ID: mdl-29020944

ABSTRACT

BACKGROUND: A sensitive, rapid and selective UHPLC-MS/MS method has been developed and validated for the quantification of Nicotine (NT) and Cotinine (CN) using Continine-d 3 as internal standard (IS) as per FDA guidelines. Sample preparation involved simple protein precipitation of 20 µL mouse plasma or brain homogenate using acetonitrile at 1:8 ratio. Mass Spectrometer was operated in positive polarity under the multiple reaction-monitoring mode using electro spray ionization technique and the transitions of m/z 163.2 â†’ 132.1, 177.2 â†’ 98.0 and 180.2 â†’ 101.2 were used to measure the NT, CN and IS, respectively. The elution of NT, CN and IS are at 1.89, 1.77 and 1.76 min, respectively. This was achieved with a gradient mobile phase consisting of 5 mM ammonium bicarbonate, acetonitrile and methanol (3:1, v/v) at a flow rate of 0.3 mL/min on a Kinetex EVO C18 column. The method was validated with a lower limit of quantitation 3.0 ng/mL in mouse plasma and brain for both the analytes. RESULTS: A linear response function was established for the range of concentrations 3-200 (r > 0.995) for NT and 3-600 ng/mL (r > 0.995) for CN. The intra- and inter-day precision values met the acceptance criteria. NT and CN are stable in the battery of stability studies viz., stock solution, bench-top and auto-sampler. CONCLUSION: This method was successfully utilized to validate a newly developed preclinical smoking model in mice.


Subject(s)
Cotinine , Nicotine , Smoking , Tandem Mass Spectrometry , Animals , Brain/enzymology , Brain/metabolism , Chromatography, High Pressure Liquid/methods , Cotinine/blood , Cotinine/cerebrospinal fluid , Mice , Nicotine/blood , Nicotine/cerebrospinal fluid , Reproducibility of Results , Sensitivity and Specificity , Smoking/physiopathology , Tandem Mass Spectrometry/methods
19.
J Pharmacol Exp Ther ; 363(3): 377-393, 2017 12.
Article in English | MEDLINE | ID: mdl-28954811

ABSTRACT

(R)-3-((3S,4S)-3-fluoro-4-(4-hydroxyphenyl)piperidin-1-yl)-1-(4-methylbenzyl)pyrrolidin-2-one (BMS-986169) and the phosphate prodrug 4-((3S,4S)-3-fluoro-1-((R)-1-(4-methylbenzyl)-2-oxopyrrolidin-3-yl)piperidin-4-yl)phenyl dihydrogen phosphate (BMS-986163) were identified from a drug discovery effort focused on the development of novel, intravenous glutamate N-methyl-d-aspartate 2B receptor (GluN2B) negative allosteric modulators (NAMs) for treatment-resistant depression (TRD). BMS-986169 showed high binding affinity for the GluN2B subunit allosteric modulatory site (Ki = 4.03-6.3 nM) and selectively inhibited GluN2B receptor function in Xenopus oocytes expressing human N-methyl-d-aspartate receptor subtypes (IC50 = 24.1 nM). BMS-986169 weakly inhibited human ether-a-go-go-related gene channel activity (IC50 = 28.4 µM) and had negligible activity in an assay panel containing 40 additional pharmacological targets. Intravenous administration of BMS-986169 or BMS-986163 dose-dependently increased GluN2B receptor occupancy and inhibited in vivo [3H](+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine ([3H]MK-801) binding, confirming target engagement and effective cleavage of the prodrug. BMS-986169 reduced immobility in the mouse forced swim test, an effect similar to intravenous ketamine treatment. Decreased novelty suppressed feeding latency, and increased ex vivo hippocampal long-term potentiation was also seen 24 hours after acute BMS-986163 or BMS-986169 administration. BMS-986169 did not produce ketamine-like hyperlocomotion or abnormal behaviors in mice or cynomolgus monkeys but did produce a transient working memory impairment in monkeys that was closely related to plasma exposure. Finally, BMS-986163 produced robust changes in the quantitative electroencephalogram power band distribution, a translational measure that can be used to assess pharmacodynamic activity in healthy humans. Due to the poor aqueous solubility of BMS-986169, BMS-986163 was selected as the lead GluN2B NAM candidate for further evaluation as a novel intravenous agent for TRD.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Organophosphates/therapeutic use , Piperidines/therapeutic use , Prodrugs/therapeutic use , Pyrrolidinones/therapeutic use , Receptors, N-Methyl-D-Aspartate/metabolism , Administration, Intravenous , Allosteric Regulation , Animals , Antidepressive Agents/adverse effects , Antidepressive Agents/pharmacokinetics , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Brain Waves/drug effects , Depressive Disorder, Major/physiopathology , Depressive Disorder, Major/psychology , Dissociative Disorders/chemically induced , Macaca fascicularis , Male , Memory, Short-Term/drug effects , Mice , Motor Activity/drug effects , Organophosphates/adverse effects , Organophosphates/pharmacokinetics , Piperidines/adverse effects , Piperidines/pharmacokinetics , Prodrugs/adverse effects , Prodrugs/pharmacokinetics , Pyrrolidinones/adverse effects , Pyrrolidinones/pharmacokinetics , Radioligand Assay , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Xenopus
20.
J Pharm Biomed Anal ; 142: 328-336, 2017 Aug 05.
Article in English | MEDLINE | ID: mdl-28535452

ABSTRACT

A highly sensitive, rapid and selective UHPLC-MS/MS method has been developed and validated for quantification of the propafenone (PF), 5-hydroxypropafenone (5-OHPF) and N-depropylpropafenone (N-DPF) on human dried blood spot (DBS). The assay procedure involves a solid-liquid extraction of PF, 5-OHPF and N-DPF and amlodipine (internal standard, I.S.) from dried human DBS cards using water and acetonitrile. The chromatographic resolution was achieved on a BEH C18 column using a gradient mobile phase consisting of 0.1% formic acid in water and acetonitrile with 0.1% formic acid at flow rate of 0.6mL/min. The UHPLC-MS/MS was operated under the multiple-reaction monitoring mode using electrospray ionization. Total run time of analysis was 1.1min and elution of PF, 5-OHPF, N-DPF and I.S. occurred at 0.69, 0.6, 0.68 and 0.73min, respectively. A detailed method validation was performed as per the regulatory guidelines and the standard curves found to be linear in the range of 5.11-1000ng/mL for PF and 5-OHPF and 0.51-100ng/mL for N-DPF with a correlation coefficient of ≥0.99 for all the drugs. The intra- and inter-day accuracies were in the range of 95.6-107 and 93.5-103; 93.4-106 and 96.3-107 and 87.9-103 and 96.5-102%, for PF, 5-OHPF and N-DPF, respectively. The intra- and inter-day precisions were in the range of 2.50-5.52 and 3.38-5.18; 2.16-6.34 and 3.23-4.94 and 2.63-7.55 and 1.56-10.2%, for PF, 5-OHPF and N-DPF, respectively. The validated assay method was successfully applied to a pharmacokinetic study in humans. The key pharmacokinetic parameters AUC0-∞ and Cmax were 6057±1526, 2002±515 and 525±202 ng*h/mL and 653±183, 295±37.5 and 68.4±13.6ng/mL for PF, 5-OHPF and N-DPF, respectively.


Subject(s)
Dried Blood Spot Testing , Chromatography, High Pressure Liquid , Humans , Propafenone/analogs & derivatives , Reproducibility of Results , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...