Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Pharmacol Exp Ther ; 379(3): 386-399, 2021 12.
Article in English | MEDLINE | ID: mdl-34535564

ABSTRACT

Ozanimod, a sphingosine 1-phosphate (S1P) receptor modulator that binds with high affinity selectively to S1P receptor subtypes 1 (S1P1) and 5 (S1P5), is approved for the treatment of relapsing multiple sclerosis (MS) in multiple countries. Ozanimod profiling revealed a species difference in its potency for S1P5 in mouse, rat, and canine compared with that for human and monkey. Site-directed mutagenesis identified amino acid alanine at position 120 to be responsible for loss of activity for mouse, rat, and canine S1P5, and mutation back to threonine as in human/monkey S1P5 restored activity. Radioligand binding analysis performed with mouse S1P5 confirmed the potency loss is a consequence of a loss of affinity of ozanimod for mouse S1P5 and was restored with mutation of alanine 120 to threonine. Study of ozanimod in preclinical mouse models of MS can now determine the S1P receptor(s) responsible for observed efficacies with receptor engagement as measured using pharmacokinetic exposures of free drug. Hence, in the experimental autoimmune encephalomyelitis model, ozanimod exposures sufficient to engage S1P1, but not S1P5, resulted in reduced circulating lymphocytes, disease scores, and body weight loss; reduced inflammation, demyelination, and apoptotic cell counts in the spinal cord; and reduced circulating levels of the neuronal degeneration marker, neurofilament light. In the demyelinating cuprizone model, ozanimod prevented axonal degradation and myelin loss during toxin challenge but did not facilitate enhanced remyelination after intoxication. Since free drug levels in this model only engaged S1P1, we concluded that S1P1 activation is neuroprotective but does not appear to affect remyelination. SIGNIFICANCE STATEMENT: Ozanimod, a selective modulator of human sphingisone 1-phosphate receptor subtypes 1 and 5 (S1P1/5), displays reduced potency for rodent and dog S1P5 compared with human, which results from mutation of threonine to alanine at position 120. Ozanimod can thus be used as a selective S1P1 agonist in mouse models of multiple sclerosis to define efficacies driven by S1P1 but not S1P5. Based on readouts for experimental autoimmune encephalomyelitis and cuprizone intoxication, S1P1 modulation is neuroprotective, but S1P5 activity may be required for remyelination.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Indans/metabolism , Multiple Sclerosis/metabolism , Oxadiazoles/metabolism , Sphingosine 1 Phosphate Receptor Modulators/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Cricetulus , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Humans , Indans/pharmacology , Indans/therapeutic use , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/genetics , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Rats , Species Specificity , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , Sphingosine 1 Phosphate Receptor Modulators/therapeutic use , Sphingosine-1-Phosphate Receptors/chemistry , Sphingosine-1-Phosphate Receptors/genetics
2.
Drug Metab Dispos ; 49(5): 405-419, 2021 05.
Article in English | MEDLINE | ID: mdl-33674268

ABSTRACT

Ozanimod is approved for the treatment of relapsing forms of multiple sclerosis. Absorption, metabolism, and excretion of ozanimod were investigated after a single oral dose of 1.0 mg [14C]ozanimod hydrochloride to six healthy subjects. In vitro experiments were conducted to understand the metabolic pathways and enzymes involved in the metabolism of ozanimod and its active metabolites. The total mean recovery of the administered radioactivity was ∼63%, with ∼26% and ∼37% recovered from urine and feces, respectively. Based on exposure, the major circulating components were active metabolite CC112273 and inactive metabolite RP101124, which together accounted for 50% of the circulating total radioactivity exposure, whereas ozanimod accounted for 6.7% of the total radioactive exposure. Ozanimod was extensively metabolized, with 14 metabolites identified, including two major active metabolites (CC112273 and CC1084037) and one major inactive metabolite (RP101124) in circulation. Ozanimod is metabolized by three primary pathways, including aldehyde dehydrogenase and alcohol dehydrogenase, cytochrome P450 isoforms 3A4 and 1A1, and reductive metabolism by gut microflora. The primary metabolite RP101075 is further metabolized to form major active metabolite CC112273 by monoamine oxidase B, which further undergoes reduction by carbonyl reductases to form CC1084037 or CYP2C8-mediated oxidation to form RP101509. CC1084037 is oxidized rapidly to form CC112273 by aldo-keto reductase 1C1/1C2 and/or 3ß- and 11ß-hydroxysteroid dehydrogenase, and this reversible oxidoreduction between two active metabolites favors CC112273. The ozanimod example illustrates the need for conducting timely radiolabeled human absorption, distribution, metabolism, and excretion studies for characterization of disproportionate metabolites and assessment of exposure coverage during drug development. SIGNIFICANCE STATEMENT: Absorption, metabolism, and excretion of ozanimod were characterized in humans, and the enzymes involved in complex metabolism were elucidated. Disproportionate metabolites were identified, and the activity of these metabolites was determined.


Subject(s)
Indans/administration & dosage , Indans/metabolism , Oxadiazoles/administration & dosage , Oxadiazoles/metabolism , Sphingosine 1 Phosphate Receptor Modulators/administration & dosage , Sphingosine 1 Phosphate Receptor Modulators/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Administration, Oral , Adult , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Male , Middle Aged
3.
Headache ; 59(6): 951-970, 2019 06.
Article in English | MEDLINE | ID: mdl-31020659

ABSTRACT

Several lines of evidence pointed to an important role for CGRP in migraine. These included the anatomic colocalization of CGRP and its receptor in sensory fibers innervating pain-producing meningeal blood vessels, its release by trigeminal stimulation, the observation of elevated CGRP in the cranial circulation during migraine with normalization concomitant with headache relief by sumatriptan, and translational studies with intravenous (IV) CGRP that evoked migraine only in migraineurs. The development of small molecule CGRP receptor antagonists (CGRP-RAs) that showed clinical antimigraine efficacy acutely and prophylactically in randomized placebo-controlled clinical trials subsequently gave definitive pharmacological proof of the importance of CGRP in migraine. More recently, CGRP target engagement imaging studies using a CGRP receptor PET ligand [11 C]MK-4232 demonstrated that there was no brain CGRP receptor occupancy at clinically effective antimigraine doses of telcagepant, a prototypic CGRP-RA. Taken together, these data indicated that (1) the therapeutic site of action of the CGRP-RAs was peripheral not central; (2) that IV CGRP had most likely evoked migraine through an action at sites outside the blood-brain barrier; and (3) that migraine pain was therefore, at least in part, peripheral in origin. The evolution of CGRP migraine science gave impetus to the development of peripherally acting drugs that could modulate CGRP chronically to prevent frequent episodic and chronic migraine. Large molecule biologic antibody (mAb) approaches that are given subcutaneously to neutralize circulating CGRP peptide (fremanezumab, galcanezumab) or block CGRP receptors (erenumab) have shown consistent efficacy and tolerability in multicenter migraine prevention trials and are now approved for clinical use. Eptinezumab, a CGRP neutralizing antibody given IV, shows promise in late stage clinical development. Recently, orally administered next-generation small molecule CGRP-RAs have been shown to have safety and efficacy in acute treatment (ubrogepant and rimegepant) and prevention (atogepant) of migraine, giving additional CGRP-based therapeutic options for migraine patients.


Subject(s)
Calcitonin Gene-Related Peptide Receptor Antagonists/therapeutic use , Calcitonin Gene-Related Peptide/antagonists & inhibitors , Calcitonin Gene-Related Peptide/metabolism , Migraine Disorders/drug therapy , Migraine Disorders/metabolism , Receptors, Calcitonin Gene-Related Peptide/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Calcitonin Gene-Related Peptide Receptor Antagonists/pharmacology , Dipeptides/pharmacology , Dipeptides/therapeutic use , Humans , Piperazines , Quinazolines/pharmacology , Quinazolines/therapeutic use , Spiro Compounds/pharmacology , Spiro Compounds/therapeutic use
4.
Neuroimage ; 106: 364-72, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25498426

ABSTRACT

Cerebral blood volume (CBV) fMRI with superparamagnetic iron oxide nanoparticles (USPIO) as contrast agent was used to investigate the odorant-induced olfaction in anesthetized rhesus monkeys. fMRI data were acquired in 24 axial slices covering the entire brain, with isoamyl-acetate as the odor stimulant. For each experiment, multiple fMRI measurements were made during a 1- or 2-h period, with each measurement consisting of a baseline period, a stimulation period, and a recovery period. Three different stimulation paradigms with a stimulation period of 1 min, 2 min, or 8 min, respectively, were used to study the olfactory responses in the olfactory bulb (OB). Odorant-induced CBV increases were observed in the OB of each individual monkey. The spatial and temporal activation patterns were reproducible within and between animals. The sensitivity of CBV fMRI in OB was comparable with the sensitivities reported in previous animal fMRI studies. The CBV responses during the 1-min, 2-min, or 8-min odor stimulation period were relatively stable, and did not show attenuation. The amplitudes of CBV response to the repeated stimuli during the 1- or 2-h period were also stable. The stable CBV response in the OB to both continuous and repeated odor stimuli suggests that the OB may not play a major role in olfactory habituation. The technical approach described in this report can enable more extensive fMRI studies of olfactory processing in OB of both humans and non-human primates.


Subject(s)
Brain Mapping/methods , Habituation, Psychophysiologic/physiology , Magnetic Resonance Imaging/methods , Olfactory Bulb/physiology , Olfactory Perception/physiology , Smell/physiology , Animals , Blood Volume/physiology , Cerebrovascular Circulation/physiology , Contrast Media , Female , Ferric Compounds , Macaca mulatta , Nanoparticles , Odorants , Olfactory Bulb/blood supply , Oxygen/blood
5.
Neurobiol Dis ; 74: 137-43, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25447225

ABSTRACT

OBJECTIVE: To examine the effect of the orexinergic blockade with a dual orexin receptor antagonist (DORA) on experimental models of peripheral and central trigeminal as well as cortical activation relevant to migraine and migraine aura. METHODS: In this study we used a precursor of suvorexant, a dual orexin receptor antagonist #12 (DORA-12) in established experimental in vivo models of dural trigeminovascular nociception in rat. Neurogenic dural vasodilation and electrophysiological recordings of second order trigeminocervical neurons were used to study trigeminal nociceptive mechanisms directly. KCl-evoked cortical spreading depression was also used as a surrogate for migraine aura. RESULTS: Neurogenically-induced vasodilation of the middle meningeal artery, caused by nociceptive activation of peripheral afferent projections of the trigeminal nerve, was attenuated by intravenous DORA-12 (1 mgkg(-1)). Second-order trigeminocervical complex neuronal activity was significantly inhibited by intravenous DORA-12 (1 mgkg(-1)). DORA-12 significantly reduced susceptibility to KCl-evoked cortical spreading depression. CONCLUSION: The study provides the first direct evidence, that simultaneous antagonism on both orexin receptors is able to attenuate trigeminal nociceptive activity as well as to induce an elevation of the threshold for the induction of a cortical spreading depression (CSD). In the clinical context, these data imply that targeting the hypothalamic orexinergic system may offer an entirely novel mechanism for the preventive treatment of migraine with and without aura.


Subject(s)
Azepines/pharmacology , Benzimidazoles/pharmacology , Migraine Disorders/drug therapy , Migraine Disorders/physiopathology , Orexin Receptor Antagonists/pharmacology , Orexin Receptors/metabolism , Animals , Azepines/chemistry , Benzimidazoles/chemistry , Central Nervous System Agents/pharmacology , Cortical Spreading Depression/drug effects , Cortical Spreading Depression/physiology , Disease Models, Animal , Electric Stimulation , Male , Microelectrodes , Nociception/drug effects , Nociception/physiology , Orexin Receptor Antagonists/chemistry , Potassium Chloride/pharmacology , Rats, Sprague-Dawley , Trigeminal Nerve/drug effects , Trigeminal Nerve/physiology , Vasodilation/drug effects , Vasodilation/physiology
6.
J Magn Reson Imaging ; 41(6): 1622-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25168165

ABSTRACT

BACKGROUND: To characterize regional kidney sodium response by MRI following NKCC2 inhibition. METHODS: Regional renal sodium signals were monitored noninvasively using (23) Na-MRI at 9.4T with a temporal resolution of 1.5 min in anesthetized rats (N = 14). A mild NKCC2 inhibition was induced using a slow intravenous furosemide infusion. Time course of sodium signal was modeled as an exponential transient with a single characteristic time constant. RESULTS: Under normal physiological conditions, the renal sodium signals in medullary and cortical regions were stable and found to respond differently to furosemide challenge. Furosemide infusion at 1.2 mg/kg/h (N = 7) increased sodium signal in the cortex by 40 ± 6% (P < 7 × 10(-5) ) whereas decreased in the medulla by 29 ± 2% (P < 3 × 10(-6) ) with different temporal kinetics. The characteristic time constants of the change were determined to be: 8 ± 2 and 70 ± 10 min for medulla and cortex. Also, the medullary change occurred 9(±3) times faster than cortical independent of furosemide infusion rate up to 35 mg/kg/h. CONCLUSION: The pharmacological effects in terms of regional kidney sodium signal changes induced by NKCC2 inhibition are region-specific and highly predictable. Using noninvasive sodium MRI, we obtained regional renal sodium kinetics data sets in response to a low dose furosemide infusion in normal rats.


Subject(s)
Diuretics/pharmacology , Furosemide/pharmacology , Kidney Cortex/drug effects , Kidney Medulla/drug effects , Magnetic Resonance Imaging/methods , Sodium/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley
7.
Neuroimage ; 84: 724-32, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24064074

ABSTRACT

fMRI can objectively measure pain-related neural activities in humans and animals, providing a valuable tool for studying the mechanisms of nociception and for developing new analgesics. However, due to its extreme sensitivity to subject motion, pain fMRI studies are performed in animals that are immobilized, typically with anesthesia. Since anesthesia could confound the nociceptive processes, it is unknown how well nociceptive-related neural activities measured by fMRI in anesthetized animals correlate with nociceptive behaviors in conscious animals. The threshold to vocalization (VT) in response to an increasing noxious electrical stimulus (NES) was implemented in conscious rats as a behavioral measure of nociception. The antinociceptive effect of systemic (intravenous infusion) lidocaine on NES-induced fMRI signals in anesthetized rats was compared with the corresponding VT in conscious rats. Lidocaine infusion increased VT and suppressed the NES-induced fMRI signals in most activated brain regions. The temporal characteristics of the nociception signal by fMRI and by VT in response to lidocaine infusion were highly correlated with each other, and with the pharmacokinetics (PK) of lidocaine. These results indicate that the fMRI activations in these regions may be used as biomarkers of acute nociception in anesthetized rats. Interestingly, systemic lidocaine had no effect on NES-induced fMRI activations in the primary somatosensory cortex (S1), a result that warrants further investigation.


Subject(s)
Brain/physiology , Pain Measurement/methods , Pain , Anesthesia, General , Anesthetics, Local/pharmacology , Animals , Brain/drug effects , Brain Mapping , Consciousness , Image Processing, Computer-Assisted , Lidocaine/pharmacology , Magnetic Resonance Imaging , Pain/physiopathology , Rats , Rats, Sprague-Dawley , Vocalization, Animal
8.
Neurobiol Dis ; 61: 32-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24055214

ABSTRACT

The goal of any early central nervous system (CNS) drug development program is always to test the mechanism and not the molecule in order to support additional research investments in late phase clinical trials. Confirmation that drugs reach their targets using translational positron emission tomography (PET) imaging markers of engagement is central to successful clinical proof-of-concept testing and has become an important feature of most neuropsychiatric drug development programs. CNS PET imaging can also play an important role in the clinical investigation of the neuropharmacological basis of psychiatric disease and the optimization of drug therapy.


Subject(s)
Central Nervous System Agents , Drug Discovery/methods , Positron-Emission Tomography/methods , Translational Research, Biomedical , Central Nervous System Agents/therapeutic use , Humans , Mental Disorders/diagnosis , Mental Disorders/drug therapy
9.
Hum Psychopharmacol ; 29(6): 568-77, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25330122

ABSTRACT

OBJECTIVE: Aprepitant is a neurokinin 1 receptor antagonist approved for prevention of chemotherapy-induced and post-operative nausea and vomiting. Early studies demonstrated promising antidepressant activity as monotherapy, although this was unsupported by subsequent phase 3 trials. This phase 2 study evaluated whether aprepitant potentiated the antidepressant effects of paroxetine. METHODS: Outpatients with major depressive disorder were randomized to aprepitant 200 mg + paroxetine 20 mg, paroxetine + placebo, or aprepitant + placebo for 6 weeks. The primary endpoint was change in HAMD-17 total score. Secondary/exploratory endpoints included changes in HAMA, CGI-S, CGI-I, and HAMD Item-1 scores at week 6. RESULTS: A total of 79, 78, and 79 patients received aprepitant + paroxetine, paroxetine + placebo, and aprepitant + placebo, respectively. At week 6, mean changes in HAMD-17 were -11.0 (95% confidence interval [CI]: -12.7, -9.4), -11.7 (95% CI: -13.3, -10.0), and -9.5 (95% CI: -10.9, -8.1), respectively. Pairwise comparisons of HAMD-17 change with combination therapy versus paroxetine alone demonstrated no significant difference (p = 0.567). Changes in CGI-S, CGI-I, and HAMD Item-1 scores were also comparable, although there was a greater reduction in anxiety (HAMA) with paroxetine alone than aprepitant + paroxetine (p = 0.045). Adverse events were generally more common with the combination than either monotherapy. CONCLUSION: Concomitant use of aprepitant + paroxetine for 6 weeks did not provide greater antidepressant benefit compared with paroxetine + placebo in patients with major depression.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Morpholines/therapeutic use , Neurokinin-1 Receptor Antagonists/therapeutic use , Paroxetine/therapeutic use , Selective Serotonin Reuptake Inhibitors/therapeutic use , Adult , Antidepressive Agents/adverse effects , Antidepressive Agents/pharmacokinetics , Aprepitant , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Male , Morpholines/adverse effects , Morpholines/pharmacokinetics , Neurokinin-1 Receptor Antagonists/adverse effects , Neurokinin-1 Receptor Antagonists/pharmacokinetics , Paroxetine/adverse effects , Paroxetine/pharmacokinetics , Psychiatric Status Rating Scales , Selective Serotonin Reuptake Inhibitors/adverse effects , Selective Serotonin Reuptake Inhibitors/pharmacokinetics , Treatment Outcome
10.
J Labelled Comp Radiopharm ; 57(10): 611-4, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25156811

ABSTRACT

The fluorine-18-labeled positron emission tomography (PET) radiotracer [(18) F]MK-9470 is a selective, high affinity inverse agonist that has been used to image the cannabinoid receptor type 1 in human brain in healthy and disease states. This report describes a simplified, one-step [(18) F]radiofluorination approach using a GE TRACERlab FXFN module for the routine production of this tracer. The one-step synthesis, by [(18) F]fluoride displacement of a primary tosylate precursor, gives a six-fold increase in yield over the previous two-step method employing O-alkylation of a phenol precursor with 1,2-[(18) F]fluorobromoethane. The average radiochemical yield of [(18) F]MK-9470 using the one-step method was 30.3 ± 11.7% (n = 12), with specific activity in excess of 6 Ci/µmol and radiochemical purity of 97.2 ± 1.5% (n = 12), in less than 60 min. This simplified, high yielding, automated process was validated for routine GMP production of [(18) F]MK-9470 for clinical studies.


Subject(s)
Pyridines/chemical synthesis , Radiopharmaceuticals/chemical synthesis
11.
Cell Metab ; 7(1): 68-78, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18177726

ABSTRACT

Cannabinoid 1 receptor (CB1R) inverse agonists are emerging as a potential obesity therapy. However, the physiological mechanisms by which these agents modulate human energy balance are incompletely elucidated. Here, we describe a comprehensive clinical research study of taranabant, a structurally novel acyclic CB1R inverse agonist. Positron emission tomography imaging using the selective CB1R tracer [(18)F]MK-9470 confirmed central nervous system receptor occupancy levels ( approximately 10%-40%) associated with energy balance/weight-loss effects in animals. In a 12-week weight-loss study, taranabant induced statistically significant weight loss compared to placebo in obese subjects over the entire range of evaluated doses (0.5, 2, 4, and 6 mg once per day) (p < 0.001). Taranabant treatment was associated with dose-related increased incidence of clinical adverse events, including mild to moderate gastrointestinal and psychiatric effects. Mechanism-of-action studies suggest that engagement of the CB1R by taranabant leads to weight loss by reducing food intake and increasing energy expenditure and fat oxidation.


Subject(s)
Amides/pharmacology , Energy Intake/drug effects , Energy Metabolism/drug effects , Pyridines/pharmacology , Receptor, Cannabinoid, CB1/agonists , Weight Loss/drug effects , Adult , Aged , Amides/therapeutic use , Brain/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Fats/metabolism , Humans , Middle Aged , Positron-Emission Tomography , Pyridines/therapeutic use
12.
Neuroimage ; 68: 1-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23238431

ABSTRACT

Antagonism of the central opioid receptor like-1 receptor (ORL1) has been implicated in cognition, and has been a focus of drug discovery efforts to ameliorate the cognitive deficits that remain during the stable treatment of schizophrenia with current antipsychotics. In order to facilitate dose selection for phase II clinical testing an ORL1-specific PET tracer was developed to determine drug plasma concentration versus occupancy relationships in order to ensure that the doses selected and the degree of target engagement were sufficient to ensure adequate proof of concept testing. MK-0911 is a selective, high affinity antagonist for the ORL1 receptor radiolabeled with high specific activity (18)F for positron emission tomography (PET) studies. Evaluation of [(18)F]MK-0911 in rhesus monkey PET studies showed a pattern of brain uptake which was consistent with the known distribution of ORL1. In vitro autoradiography with [(18)F]MK-0911 in rhesus monkey and human brain tissue slices showed a regional distribution that was consistent with in vivo imaging results in monkey. Pre-treatment of rhesus monkeys with high doses of structurally diverse ORL1 antagonists MK-0584, MK-0337, or MK-5757 achieved blockade of [(18)F]MK-0911 in all gray matter regions. Baseline PET studies with [(18)F]MK-0911 in healthy human subjects showed tracer distribution and kinetics similar to that observed in rhesus monkey. Quantification of [(18)F]MK-0911 uptake in repeat human baseline PET studies showed a test-retest variability in volume of distribution (V(T)) averaging 3% across brain regions. Humans dosed orally with MK-5757 showed reduced [(18)F]MK-0911 tracer concentration in brain proportional with MK-5757 dose and plasma level. [(18)F]MK-0911 was useful for determining MK-5757-induced receptor occupancy of ORL1 to guide MK-5757 dose-selection for clinical proof-of-concept studies. Additionally, [(18)F]MK-0911 may be a useful tool for studying the pharmacology of ORL1 in various human populations and disease states.


Subject(s)
Benzimidazoles/pharmacokinetics , Brain/diagnostic imaging , Fluorine Radioisotopes/pharmacokinetics , Piperidines/pharmacokinetics , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Receptors, Opioid/metabolism , Adult , Animals , Benzimidazoles/chemistry , Brain/metabolism , Fluorine Radioisotopes/chemistry , Humans , Macaca mulatta , Male , Middle Aged , Piperidines/chemistry , Radiopharmaceuticals/chemistry , Tissue Distribution , Young Adult , Nociceptin Receptor
13.
J Pharmacol Exp Ther ; 345(1): 41-51, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23370795

ABSTRACT

Pharmacological magnetic resonance imaging (phMRI) is one method by which a drug's pharmacodynamic effects in the brain can be assessed. Although phMRI has been frequently used in preclinical and clinical settings, the extent to which a phMRI signature for a compound translates between rodents and humans has not been systematically examined. In the current investigation, we aimed to build on recent clinical work in which the functional response to 0.1 and 0.2 mg/70 kg i.v. buprenorphine (partial µ-opioid receptor agonist) was measured in healthy humans. Here, we measured the phMRI response to 0.04 and 0.1 mg/kg i.v. buprenorphine in conscious, naive rats to establish the parallelism of the phMRI signature of buprenorphine across species. PhMRI of 0.04 and 0.1 mg/kg i.v. buprenorphine yielded dose-dependent activation in a brain network composed of the somatosensory cortex, cingulate, insula, striatum, thalamus, periaqueductal gray, and cerebellum. Similar dose-dependent phMRI activation was observed in the human phMRI studies. These observations indicate an overall preservation of pharmacodynamic responses to buprenorphine between conscious, naive rodents and healthy human subjects, particularly in brain regions implicated in pain and analgesia. This investigation further demonstrates the usefulness of phMRI as a translational tool in neuroscience research that can provide mechanistic insight and guide dose selection in drug development.


Subject(s)
Analgesics, Opioid/pharmacology , Brain/drug effects , Buprenorphine/pharmacology , Magnetic Resonance Imaging , Neurosciences , Animals , Brain Mapping/methods , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Infusions, Intravenous , Magnetic Resonance Imaging/instrumentation , Magnetic Resonance Imaging/methods , Male , Neurosciences/instrumentation , Neurosciences/methods , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Species Specificity
14.
J Pharmacol Exp Ther ; 347(2): 478-86, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23975906

ABSTRACT

Calcitonin gene-related peptide (CGRP) is a potent neuropeptide whose agonist interaction with the CGRP receptor (CGRP-R) in the periphery promotes vasodilation, neurogenic inflammation and trigeminovascular sensory activation. This process is implicated in the cause of migraine headaches, and CGRP-R antagonists in clinical development have proven effective in treating migraine-related pain in humans. CGRP-R is expressed on blood vessel smooth muscle and sensory trigeminal neurons and fibers in the periphery as well as in the central nervous system. However, it is not clear what role the inhibition of central CGRP-R plays in migraine pain relief. To this end, the CGRP-R positron emission tomography (PET) tracer [(11)C]MK-4232 (2-[(8R)-8-(3,5-difluorophenyl)-6,8-[6-(11)C]dimethyl-10-oxo-6,9-diazaspiro[4.5]decan-9-yl]-N-[(2R)-2'-oxospiro[1,3-dihydroindene-2,3'-1H-pyrrolo[2,3-b]pyridine]-5-yl]acetamide) was discovered and developed for use in clinical PET studies. In rhesus monkeys and humans, [(11)C]MK-4232 displayed rapid brain uptake and a regional brain distribution consistent with the known distribution of CGRP-R. Monkey PET studies with [(11)C]MK-4232 after intravenous dosing with CGRP-R antagonists validated the ability of [(11)C]MK-4232 to detect changes in CGRP-R occupancy in proportion to drug plasma concentration. Application of [(11)C]MK-4232 in human PET studies revealed that telcagepant achieved only low receptor occupancy at an efficacious dose (140 mg PO). Therefore, it is unlikely that antagonism of central CGRP-R is required for migraine efficacy. However, it is not known whether high central CGRP-R antagonism may provide additional therapeutic benefit.


Subject(s)
Acetanilides/pharmacokinetics , Analgesics/pharmacokinetics , Azepines/pharmacokinetics , Brain/metabolism , Calcitonin Gene-Related Peptide Receptor Antagonists , Imidazoles/pharmacokinetics , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Spiro Compounds/pharmacokinetics , Acetanilides/chemistry , Adult , Analgesics/therapeutic use , Animals , Azepines/therapeutic use , Brain/diagnostic imaging , Carbon Radioisotopes , Female , Humans , Imidazoles/therapeutic use , Macaca mulatta , Male , Middle Aged , Migraine Disorders/drug therapy , Migraine Disorders/metabolism , Molecular Structure , Protein Binding , Radiopharmaceuticals/chemistry , Species Specificity , Spiro Compounds/chemistry , Tissue Distribution , Young Adult
15.
Int J Neuropsychopharmacol ; 16(1): 1-11, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22433185

ABSTRACT

Preclinical studies suggest that substance P acting at neurokinin 1 (NK1) receptors may be involved in stress responses and NK1 receptor antagonists show activity in tests of anxiety. These data raise the possibility that NK1 receptor antagonists could be potential anxiolytic treatments in humans. We evaluated this hypothesis clinically using the NK1 antagonist L-759274. This is a randomized, double-blind, placebo- and active-controlled, multicentre, proof-of-concept trial. Patients with generalized anxiety disorder were randomized 1:1:1 to 6 wk of treatment with 40 mg L-759274 (n = 73), 1-6 mg lorazepam (n = 69) or placebo (n = 71). Efficacy was assessed using the Hamilton Anxiety Scale (HAMA). A positron emission tomography (PET) study was also performed in 16 healthy subjects to determine the relationship between NK1 receptor occupancy and plasma levels of L-759274 to verify adequate target engagement by the doses tested during the clinical trial. No statistically significant difference in mean change from baseline HAMA score at 6 wk was seen for L-759274 vs. placebo [difference = 1.0 (95% confidence intervals (CI) -1.2 to 3.2), p = 0.359] whereas the lorazepam group did show a significant improvement vs. placebo (difference = -2.7, 95% CI -5.0 to -0.4, p = 0.020) and L-759274 (difference = 3.7, 95% CI 1.5-6.0, p = 0.001]. Results from the PET study indicated that the L-759274 dosing regimen used in the clinical trial likely provided high levels of NK1 receptor occupancy (>90%), supporting the view that it was an adequate proof-of-concept trial. The NK1 receptor antagonist L-759274 does not appear to be efficacious for the treatment of generalized anxiety disorder.


Subject(s)
Anti-Anxiety Agents/therapeutic use , Anxiety Disorders/diagnostic imaging , Anxiety Disorders/drug therapy , Neurokinin-1 Receptor Antagonists , Adult , Anti-Anxiety Agents/pharmacology , Anxiety Disorders/psychology , Double-Blind Method , Female , Humans , Lorazepam/pharmacology , Lorazepam/therapeutic use , Male , Middle Aged , Positron-Emission Tomography/methods , Treatment Outcome
16.
Curr Pain Headache Rep ; 17(10): 369, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24037443

ABSTRACT

The relationship between sleep and migraine headaches is complex. Changes in sleep patterns can trigger migraine attacks, and sleep disorders may be associated with increased migraine frequency. Furthermore, migraine patients and their doctors very consistently report that sleep relieves already established migraine attacks. Herein we will try to answer the question, "Why does sleep stop migraine?" Since evidence for this relationship is largely based on empirical clinical observation, we will not provide a clinical review of the association. Instead, we will focus on the pathophysiology of migraine attacks and its intersections with sleep biology.


Subject(s)
Migraine Disorders/physiopathology , Sleep/physiology , Humans
17.
Eur J Pharmacol ; 941: 175442, 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36470447

ABSTRACT

Ozanimod is approved in multiple countries for the treatment of adults with either relapsing multiple sclerosis or moderately to severely active ulcerative colitis. Ozanimod is metabolized in humans to form seven active plasma metabolites, including two major active metabolites CC112273 and CC1084037, and an inactive metabolite. Here, the binding and activity of ozanimod and its metabolites across human sphingosine 1-phosphate receptors were determined. Binding affinity was assessed in Chinese hamster ovary cell membranes expressing recombinant human sphingosine 1-phosphate receptors 1 and 5 via competitive radioligand binding using tritium-labeled ozanimod; selectivity via functional potency assessment was performed using [35S]-guanosine-5'-(γ-thio)-triphosphate binding assays. Receptor internalization was assessed in human embryonic kidney 293 cells overexpressing sphingosine 1-phosphate receptor 1-green fluorescent protein and Chinese hamster ovary cells overexpressing sphingosine 1-phosphate receptor 5-hemagglutinin via fluorescence activated cell sorting. Functional activity was assessed in primary cultures of human astrocytes via phosphorylation assays. Ozanimod and its functionally active metabolites bound to the same sites within sphingosine 1-phosphate receptors 1 and 5, with metabolites displaying the same selectivity profile as ozanimod. Agonism at sphingosine 1-phosphate receptor 1 induced receptor internalization, whereas sphingosine 1-phosphate receptor 5 did not. Ozanimod, CC112273, and CC1084037 elicited functional intracellular signaling in human astrocytes, pharmacologically characterized to be mediated by sphingosine 1-phosphate receptor 1. The active plasma metabolites of ozanimod bound to sphingosine 1-phosphate receptors 1 and 5 and displayed similar pharmacologic profiles as their parent compound, likely contributing to clinical efficacy in patients with relapsing multiple sclerosis or moderately to severely active ulcerative colitis.


Subject(s)
Colitis, Ulcerative , Multiple Sclerosis , Adult , Animals , Cricetinae , Humans , Sphingosine-1-Phosphate Receptors/metabolism , Colitis, Ulcerative/drug therapy , CHO Cells , Cricetulus , Indans/pharmacology , Indans/therapeutic use , Oxadiazoles/pharmacology , Sphingosine , Multiple Sclerosis/drug therapy
18.
J Biol Chem ; 286(19): 17217-26, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21454574

ABSTRACT

Apolipoprotein (apo) E4 is the major genetic risk factor for Alzheimer disease (AD) and likely contributes to neuropathology through various pathways. Here we report that the intracellular trafficking of apoE4 is impaired in Neuro-2a cells and primary neurons, as shown by measuring fluorescence recovery after photobleaching. In Neuro-2a cells, more apoE4 than apoE3 molecules remained immobilized in the endoplasmic reticulum (ER) and the Golgi apparatus, and the lateral motility of apoE4 was significantly lower in the Golgi apparatus (but not in the ER) than that of apoE3. Likewise, the immobile fraction was larger, and the lateral motility was lower for apoE4 than apoE3 in mouse primary hippocampal neurons. ApoE4 with the R61T mutation, which abolishes apoE4 domain interaction, was less immobilized, and its lateral motility was comparable with that of apoE3. The trafficking impairment of apoE4 was also rescued by disrupting domain interaction with the small-molecule structure correctors GIND25 and PH002. PH002 also rescued apoE4-induced impairments of neurite outgrowth in Neuro-2a cells and dendritic spine development in primary neurons. ApoE4 did not affect trafficking of amyloid precursor protein, another AD-related protein, through the secretory pathway. Thus, domain interaction renders more newly synthesized apoE4 molecules immobile and slows their trafficking along the secretory pathway. Correcting the pathological structure of apoE4 by disrupting domain interaction is a potential therapeutic approach to treat or prevent AD related to apoE4.


Subject(s)
Apolipoprotein E4/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Biological Transport , Cell Line , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Fluorescence Recovery After Photobleaching , Golgi Apparatus/metabolism , Hippocampus/cytology , Humans , Mice , Models, Biological , Mutation , Neurons/metabolism
19.
Neuroimage ; 59(2): 1168-79, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-21856430

ABSTRACT

This study aims to identify fMRI signatures of nociceptive processing in whole brain of anesthetized rats during noxious electrical stimulation (NES) and noxious mechanical stimulation (NMS) of paw. Activation patterns for NES were mapped with blood oxygen level dependent (BOLD) and cerebral blood volume (CBV) fMRI, respectively, to investigate the spatially-dependent hemodynamic responses during nociception processing. A systematic evaluation of fMRI responses to varying frequencies of electrical stimulus was carried out to optimize the NES protocol. Both BOLD and CBV fMRI showed widespread activations, but with different spatial characteristics. While BOLD and CBV showed well-localized activations in ipsilateral dorsal column nucleus, contralateral primary somatosensory cortex (S1), and bilateral caudate putamen (CPu), CBV fMRI showed additional bilateral activations in the regions of pons, midbrain and thalamus compared to BOLD fMRI. CBV fMRI that offers higher sensitivity compared to BOLD was then used to compare the nociception processing during NES and NMS in the same animal. The activations in most regions were similar. In the medulla, however, NES induced a robust activation in the ipsilateral dorsal column nucleus while NMS showed no activation. This study demonstrates that (1) the hemodynamic response to nociception is spatial-dependent; (2) the widespread activations during nociception in CBV fMRI are similar to what have been observed in (14)C-2-deoxyglucose (2DG) autoradiography and PET; (3) the bilateral activations in the brain originate from the divergence of neural responses at supraspinal level; and (4) the similarity of activation patterns suggests that nociceptive processing in rats is similar during NES and NMS.


Subject(s)
Brain/physiopathology , Cerebrovascular Circulation , Electric Stimulation , Magnetic Resonance Imaging/methods , Nerve Net/physiopathology , Pain/physiopathology , Physical Stimulation , Animals , Blood Flow Velocity , Blood Volume , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity
20.
Neuroimage ; 59(4): 3762-73, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22119647

ABSTRACT

Buprenorphine (BUP) is a partial agonist at µ-, δ- and ORL1 (opioid receptor-like)/nociceptin receptors and antagonist at the κ-opioid receptor site. BUP is known to have both analgesic as well as antihyperalgesic effects via its central activity, and is used in the treatment of moderate to severe chronic pain conditions. Recently, it was shown that intravenous (IV) administration of 0.2mg/70 kg BUP modulates the blood oxygenation level-dependent (BOLD) functional magnetic resonance imaging (fMRI) response to acute noxious stimuli in healthy human subjects. The present study extends these observations by investigating the effects of BUP dose and route of administration on central nervous system (CNS) pain circuitry. Specifically, the modulation of evoked pain BOLD responses and resting state functional connectivity was measured following IV (0.1 and 0.2mg/70 kg) and sublingual (SL) (2mg) BUP administration in healthy human subjects. While 0.1mg/70 kg IV BUP is sub-analgesic, both 0.2mg/70 kg IV BUP and 2.0mg SL BUP are analgesic doses of the drug. Evoked BOLD responses were clearly modulated in a dose-dependent manner. The analgesic doses of BUP by both routes of administration yielded a potentiation in limbic/mesolimbic circuitry and attenuation in sensorimotor/sensory-discriminative circuitry. In addition, robust decreases in functional connectivity between the putamen and the sensorimotor/sensory-discriminative structures were observed at the two analgesic doses subsequent to measuring the maximum plasma BUP concentrations (C(max)). The decreases in functional connectivity within the sensorimotor/sensory-discriminative circuitry were also observed to be dose-dependent in the IV administration cohorts. These reproducible and consistent functional CNS measures at clinically effective doses of BUP demonstrate the potential of evoked pain fMRI and resting-state functional connectivity as objective tools that can inform the process of dose selection. Such methods may be useful during early clinical phase evaluation of potential analgesics in drug development.


Subject(s)
Analgesics, Opioid/pharmacology , Brain/drug effects , Brain/physiopathology , Buprenorphine/pharmacology , Pain/drug therapy , Pain/physiopathology , Administration, Sublingual , Adult , Analgesics, Opioid/administration & dosage , Buprenorphine/administration & dosage , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Injections, Intravenous , Magnetic Resonance Imaging , Male
SELECTION OF CITATIONS
SEARCH DETAIL