Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Front Pharmacol ; 15: 1432902, 2024.
Article in English | MEDLINE | ID: mdl-39224779

ABSTRACT

Sunobinop is a novel, potent, selective partial agonist at nociceptin/orphanin FQ peptide (NOP) receptors. The primary objective of this randomized, double-blind, placebo-controlled study was to assess the next-day residual effects of an evening dose of sunobinop in healthy participants. Participants were randomized into 1 of 5 treatment sequences. Treatment consisted of 1 dose each of sunobinop 0.2, 0.6, 2, and 6 mg suspension and placebo suspension. Key pharmacodynamic (PD) measures included the digit symbol substitution test (DSST), Karolinska sleepiness scale (KSS), and body sway. The randomized safety population consisted of 25 participants. The DSST, KSS, and body sway showed dose-dependent effects following the administration of sunobinop, with no significant differences versus placebo at sunobinop doses <2 mg. At sunobinop 2 mg, PD effects were relatively small in magnitude and inconsistent. The last timepoint where significant differences between sunobinop 2 mg and placebo on the DSST, KSS, and body sway were observed was at 12 h, 16.5 h, and 13.5 h postdose, respectively. Sunobinop 6 mg resulted in larger and consistent PD effects, with significant differences from placebo at all timepoints up to 16.5-18 h postdose. Somnolence was the most frequently reported adverse event (AE), and all AEs were mild-to-moderate. No deaths occurred during the study or discontinuations due to an AE. Overall, a nighttime oral dose of sunobinop up to 2 mg was safe and generally well tolerated in healthy participants with limited next-day residual effects that were consistent with other sedative/hypnotic drugs.

2.
Clin Pharmacol Drug Dev ; 13(7): 790-800, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38476082

ABSTRACT

Sunobinop is an investigational, potent, selective partial agonist at the nociceptin/orphanin FQ peptide receptor in vitro. Three phase 1 studies were conducted to evaluate the safety, tolerability, and pharmacokinetics (PK) of escalating single- and multiple-dose administration of sunobinop in healthy participants. Study 1 was a randomized, double-blind, placebo-controlled, single-ascending dose study. Study 2 was a randomized, double-blind, placebo-controlled, multiple-ascending dose study. Study 3 was a randomized, open-label, single-dose, 4-way crossover study of oral and sublingual sunobinop comparing morning (AM) and bedtime (PM) administration. Seventy participants were included. Systemic exposure (peak plasma concentration [Cmax], area under the plasma concentration-time curve from time 0 to the time of last quantifiable concentration [AUC0-t], and area under the plasma concentration-time curve from time 0 extrapolated to infinity [AUCinf]) of sunobinop was characterized by dose proportionality from 0.6 to 2 mg and increased less than proportionally from 3 to 30 mg. The PKs of sunobinop were similar, regardless of AM or PM administration, for both the oral and sublingual formulations. The majority of absorbed sunobinop was excreted unchanged in the urine within 8 hours of dosing, thereby showing rapid elimination with no appreciable accumulation following 14 consecutive days of once-daily dosing and suggesting exclusive renal elimination. Most treatment-emergent adverse events (TEAEs) were mild in severity; 1 severe TEAE occurred and all TEAEs resolved by the end of the studies. Sunobinop was generally well-tolerated and safe across the range of doses evaluated and presents a clinical profile suitable for continued development.


Subject(s)
Area Under Curve , Cross-Over Studies , Healthy Volunteers , Humans , Male , Adult , Double-Blind Method , Female , Middle Aged , Young Adult , Administration, Oral , Dose-Response Relationship, Drug , Administration, Sublingual , Drug Administration Schedule , Nociceptin Receptor , Receptors, Opioid/metabolism , Adolescent , Morphinans/pharmacokinetics , Morphinans/administration & dosage , Morphinans/adverse effects , Naltrexone/analogs & derivatives
4.
Int J Pharm ; 618: 121658, 2022 Apr 25.
Article in English | MEDLINE | ID: mdl-35292396

ABSTRACT

Levodopa (L-DOPA) is an oral Parkinson's Disease drug that generates the active metabolite - dopamine (DA) in vivo. However, oral L-DOPA exhibits low oral bioavailability, limited brain uptake, peripheral DA-mediated side effects and its poor brain bioavailability can lead to long-term complications. Here we show that L-DOPA forms stable (for at least 5 months) 300 nm nanoparticles when encapsulated within N-palmitoyl-N-monomethyl-N,N-dimethyl-N,N,N-trimethyl-6-O-glycolchitosan (GCPQ). A nano-in-microparticle GCPQ-L-DOPA formulation (D50 = 7.2 µm), prepared by spray-drying, was stable for one month when stored at room and refrigeration temperatures and was capable of producing the original GCPQ-L-DOPA nanoparticles upon aqueous reconstitution. Nasal administration of reconstituted GCPQ-L-DOPA nanoparticles to rats resulted in significantly higher DA levels in the brain (Cmax of 94 ng g-1 above baseline levels 2 h post-dosing) when compared to nasal administration of L-DOPA alone, with DA being undetectable in the brain with the latter. Furthermore, nasal GCPQ-L-DOPA resulted in higher levels of L-DOPA in the plasma (a 17-fold increase in the Cmax, when compared to L-DOPA alone) with DA undetectable in the plasma from both formulations. These data provide evidence of effective delivery of DA to the brain with the GCPQ-L-DOPA formulation.


Subject(s)
Levodopa , Parkinson Disease , Animals , Biological Availability , Brain/metabolism , Dopamine , Levodopa/therapeutic use , Parkinson Disease/drug therapy , Rats
5.
J Neurosci ; 30(6): 2017-24, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-20147530

ABSTRACT

Endocannabinoids (eCBs) function as retrograde signaling molecules at synapses throughout the brain, regulate axonal growth and guidance during development, and drive adult neurogenesis. There remains a lack of genetic evidence as to the identity of the enzyme(s) responsible for the synthesis of eCBs in the brain. Diacylglycerol lipase-alpha (DAGLalpha) and -beta (DAGLbeta) synthesize 2-arachidonoyl-glycerol (2-AG), the most abundant eCB in the brain. However, their respective contribution to this and to eCB signaling has not been tested. In the present study, we show approximately 80% reductions in 2-AG levels in the brain and spinal cord in DAGLalpha(-/-) mice and a 50% reduction in the brain in DAGLbeta(-/-) mice. In contrast, DAGLbeta plays a more important role than DAGLalpha in regulating 2-AG levels in the liver, with a 90% reduction seen in DAGLbeta(-/-) mice. Levels of arachidonic acid decrease in parallel with 2-AG, suggesting that DAGL activity controls the steady-state levels of both lipids. In the hippocampus, the postsynaptic release of an eCB results in the transient suppression of GABA-mediated transmission at inhibitory synapses; we now show that this form of synaptic plasticity is completely lost in DAGLalpha(-/-) animals and relatively unaffected in DAGLbeta(-/-) animals. Finally, we show that the control of adult neurogenesis in the hippocampus and subventricular zone is compromised in the DAGLalpha(-/-) and/or DAGLbeta(-/-) mice. These findings provide the first evidence that DAGLalpha is the major biosynthetic enzyme for 2-AG in the nervous system and reveal an essential role for this enzyme in regulating retrograde synaptic plasticity and adult neurogenesis.


Subject(s)
Brain/metabolism , Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Lipoprotein Lipase/genetics , Animals , Arachidonic Acids/metabolism , Brain/cytology , Glycerides/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Liver/metabolism , Mice , Mice, Knockout , Neurogenesis , Neuronal Plasticity , Signal Transduction , Spinal Cord/metabolism , Synapses/physiology
6.
Mol Pharmacol ; 77(6): 953-60, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20223879

ABSTRACT

P2X5 is a member of the P2X family of ATP-gated nonselective cation channels, which exist as trimeric assemblies. P2X5 is believed to trimerize with another member of this family, P2X1. We investigated the single-nucleotide polymorphism (SNP) at the 3' splice site of exon 10 of the human P2X5 gene. As reported previously, presence of a T at the SNP location results in inclusion of exon 10 in the mature transcript, whereas exon 10 is excluded when a G is present at this location. Our genotyping of human DNA samples reveals predominance of the G-bearing allele, which was exclusively present in DNA samples from white American, Middle Eastern, and Chinese donors. Samples from African American donors were polymorphic, with the G allele more frequent. Reverse transcription-polymerase chain reaction analysis of lymphocytes demonstrated a 100% positive correlation between genotype and P2X5 transcript. Immunostaining of P2X1/P2X5 stably coexpressing cell lines showed full-length P2X5 to be expressed at the cell surface and the exon 10-deleted isoform to be cytoplasmic. Fluorometric imaging-based pharmacological characterization indicated a ligand-dependent increase in intracellular calcium in 1321N1 astrocytoma cells transiently expressing full-length P2X5 but not the exon 10-deleted isoform. Likewise, electrophysiological analysis showed robust ATP-evoked currents when full-length but not the exon 10-deleted isoform of P2X5 was expressed. Taken together, our findings indicate that most humans express only a nonfunctional isoform of P2X5, which is in stark contrast to what is seen in other vertebrate species in which P2X5 has been studied, from which only the full-length isoform is known.


Subject(s)
Exons , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Receptors, Purinergic P2/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Cell Line , Humans , Immunohistochemistry , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/physiology , Receptors, Purinergic P2/chemistry , Receptors, Purinergic P2/physiology , Receptors, Purinergic P2X5 , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
7.
Mol Pharmacol ; 78(6): 996-1003, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20855465

ABSTRACT

Endocannabinoids are lipid molecules that serve as natural ligands for the cannabinoid receptors CB1 and CB2. They modulate a diverse set of physiological processes such as pain, cognition, appetite, and emotional states, and their levels and functions are tightly regulated by enzymatic biosynthesis and degradation. 2-Arachidonoylglycerol (2-AG) is the most abundant endocannabinoid in the brain and is believed to be hydrolyzed primarily by the serine hydrolase monoacylglycerol lipase (MAGL). Although 2-AG binds and activates cannabinoid receptors in vitro, when administered in vivo, it induces only transient cannabimimetic effects as a result of its rapid catabolism. Here we show using a mouse model with a targeted disruption of the MAGL gene that MAGL is the major modulator of 2-AG hydrolysis in vivo. Mice lacking MAGL exhibit dramatically reduced 2-AG hydrolase activity and highly elevated 2-AG levels in the nervous system. A lack of MAGL activity and subsequent long-term elevation of 2-AG levels lead to desensitization of brain CB1 receptors with a significant reduction of cannabimimetic effects of CB1 agonists. Also consistent with CB1 desensitization, MAGL-deficient mice do not show alterations in neuropathic and inflammatory pain sensitivity. These findings provide the first genetic in vivo evidence that MAGL is the major regulator of 2-AG levels and signaling and reveal a pivotal role for 2-AG in modulating CB1 receptor sensitization and endocannabinoid tone.


Subject(s)
Cannabinoid Receptor Modulators/physiology , Endocannabinoids , Monoacylglycerol Lipases/metabolism , Receptor, Cannabinoid, CB1/physiology , Animals , Enzyme Activation/genetics , Enzyme Activation/physiology , Hydrolysis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monoacylglycerol Lipases/deficiency , Monoacylglycerol Lipases/physiology , Pain Measurement/methods
8.
Neurobiol Dis ; 39(2): 148-55, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20381618

ABSTRACT

Rimonabant was the first clinically marketed cannabinoid (CB)(1) receptor antagonist developed to treat obesity. Unfortunately, CB(1) receptor antagonism produced adverse psychiatric events in patients. To determine whether this occurs pre-clinically, we investigated the effects of rimonabant in rodent models of mood disorders. Chronic treatment with rimonabant increased immobility time in the rat forced swim test and reduced the consumption of sucrose-sweetened water in an assay postulated to model anhedonia. These responses were similar to the effects elicited by chronic mild stress in these behavioral models, which, taken together, are indicative of a depression-like phenotype. Additionally, chronic treatment with rimonabant produced decreases in frontal cortex serotonin levels, marked reductions in hippocampal cell proliferation, survival, and BDNF levels, and elevations in the concentrations of pro-inflammatory cytokines including interferon gamma and TNF alpha. These preclinical findings mimic clinical reports and implicate possible mechanisms responsible for the unfavorable psychiatric events reported following chronic rimonabant use.


Subject(s)
Phenotype , Piperidines/adverse effects , Pyrazoles/adverse effects , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Stress, Psychological/chemically induced , Stress, Psychological/physiopathology , Analysis of Variance , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/metabolism , Disease Models, Animal , Flow Cytometry/methods , Food Preferences/drug effects , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Immobility Response, Tonic/drug effects , Male , Microdialysis/methods , Rats , Rats, Sprague-Dawley , Rimonabant , Stress, Psychological/pathology , Sucrose/administration & dosage , Sweetening Agents/administration & dosage , Swimming/psychology
9.
BMC Neurosci ; 11: 8, 2010 Jan 20.
Article in English | MEDLINE | ID: mdl-20085661

ABSTRACT

BACKGROUND: Targeted delivery of pharmaceutical agents into selected populations of CNS (Central Nervous System) neurons is an extremely compelling goal. Currently, systemic methods are generally used for delivery of pain medications, anti-virals for treatment of dermatomal infections, anti-spasmodics, and neuroprotectants. Systemic side effects or undesirable effects on parts of the CNS that are not involved in the pathology limit efficacy and limit clinical utility for many classes of pharmaceuticals. Axonal transport from the periphery offers a possible selective route, but there has been little progress towards design of agents that can accomplish targeted delivery via this intraneural route. To achieve this goal, we developed a tripartite molecular construction concept involving an axonal transport facilitator molecule, a polymer linker, and a large number of drug molecules conjugated to the linker, then sought to evaluate its neurobiology and pharmacological behavior. RESULTS: We developed chemical synthesis methodologies for assembling these tripartite complexes using a variety of axonal transport facilitators including nerve growth factor, wheat germ agglutinin, and synthetic facilitators derived from phage display work. Loading of up to 100 drug molecules per complex was achieved. Conjugation methods were used that allowed the drugs to be released in active form inside the cell body after transport. Intramuscular and intradermal injection proved effective for introducing pharmacologically effective doses into selected populations of CNS neurons. Pharmacological efficacy with gabapentin in a paw withdrawal latency model revealed a ten fold increase in half life and a 300 fold decrease in necessary dose relative to systemic administration for gabapentin when the drug was delivered by axonal transport using the tripartite vehicle. CONCLUSION: Specific targeting of selected subpopulations of CNS neurons for drug delivery by axonal transport holds great promise. The data shown here provide a basic framework for the intraneural pharmacology of this tripartite complex. The pharmacologically efficacious drug delivery demonstrated here verify the fundamental feasibility of using axonal transport for targeted drug delivery.


Subject(s)
Axonal Transport , Drug Delivery Systems/methods , Neurons/drug effects , Amines/administration & dosage , Amines/chemistry , Amines/pharmacokinetics , Amines/pharmacology , Analgesics/administration & dosage , Analgesics/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Cell Line , Cells, Cultured , Cricetinae , Cyclohexanecarboxylic Acids/administration & dosage , Cyclohexanecarboxylic Acids/chemistry , Cyclohexanecarboxylic Acids/pharmacokinetics , Cyclohexanecarboxylic Acids/pharmacology , Dextrans/chemistry , Dextrans/pharmacology , Dose-Response Relationship, Drug , Gabapentin , Half-Life , Macaca fascicularis , Models, Neurological , Nanoparticles/chemistry , Nerve Growth Factor/chemistry , Nerve Growth Factor/pharmacokinetics , Neurons/ultrastructure , Pain/drug therapy , Polymers/chemistry , Polymers/pharmacokinetics , Polymers/pharmacology , Rabbits , Rats , Rats, Sprague-Dawley , Wheat Germ Agglutinins/chemistry , Wheat Germ Agglutinins/pharmacokinetics , gamma-Aminobutyric Acid/administration & dosage , gamma-Aminobutyric Acid/chemistry , gamma-Aminobutyric Acid/pharmacokinetics , gamma-Aminobutyric Acid/pharmacology
10.
Bioorg Med Chem Lett ; 20(5): 1555-8, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20153188

ABSTRACT

Two related series of selective norepinephrine reuptake inhibitors were synthesized based on 3,4-dihydro-1H-2,1,3-benzothiadiazine 2,2-dioxide or 3,4-dihydrosulfostyril cores, and screened for monoamine reuptake inhibition. Structure-activity relationships were determined for the series' in vitro potency and selectivity versus serotonin or dopamine transporter inhibition, and analogs based on both cores were identified as potent and selective NRIs. The 3,4-dihydrosulfostyril series was further tested for microsome stability, and compound 16j, which was optimized for both potency and stability, showed efficacy in an in vivo model of thermoregulatory dysfunction.


Subject(s)
Adrenergic Uptake Inhibitors/chemistry , Benzothiadiazines/chemistry , Cyclic S-Oxides/chemistry , Norepinephrine Plasma Membrane Transport Proteins/chemistry , Norepinephrine/metabolism , Thiazines/chemistry , Adrenergic Uptake Inhibitors/chemical synthesis , Adrenergic Uptake Inhibitors/pharmacology , Animals , Biological Transport , Cyclic S-Oxides/chemical synthesis , Cyclic S-Oxides/pharmacology , Humans , Microsomes, Liver/metabolism , Models, Animal , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Rats , Structure-Activity Relationship , Thiazines/chemical synthesis , Thiazines/pharmacology
11.
Brain Res Rev ; 60(1): 255-66, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19150370

ABSTRACT

The endocannabinoid system consists of the cannabinoid (CB) receptors, CB(1) and CB(2), the endogenous ligands anandamide (AEA, arachidonoylethanolamide) and 2-arachidonoylglycerol (2-AG), and their synthetic and metabolic machinery. The use of cannabis has been described in classical and recent literature for the treatment of pain, but the potential for psychotropic effects as a result of the activation of central CB(1) receptors places a limitation upon its use. There are, however, a number of modern approaches being undertaken to circumvent this problem, and this review represents a concise summary of these approaches, with a particular emphasis upon CB(2) receptor agonists. Selective CB(2) agonists and peripherally restricted CB(1) or CB(1)/CB(2) dual agonists are being developed for the treatment of inflammatory and neuropathic pain, as they demonstrate efficacy in a range of pain models. CB(2) receptors were originally described as being restricted to cells of immune origin, but there is evidence for their expression in human primary sensory neurons, and increased levels of CB(2) receptors reported in human peripheral nerves have been seen after injury, particularly in painful neuromas. CB(2) receptor agonists produce antinociceptive effects in models of inflammatory and nociceptive pain, and in some cases these effects involve activation of the opioid system. In addition, CB receptor agonists enhance the effect of mu-opioid receptor agonists in a variety of models of analgesia, and combinations of cannabinoids and opioids may produce synergistic effects. Antinociceptive effects of compounds blocking the metabolism of anandamide have been reported, particularly in models of inflammatory pain. There is also evidence that such compounds increase the analgesic effect of non-steroidal anti-inflammatory drugs (NSAIDs), raising the possibility that a combination of suitable agents could, by reducing the NSAID dose needed, provide an efficacious treatment strategy, while minimizing the potential for NSAID-induced gastrointestinal and cardiovascular disturbances. Other potential "partners" for endocannabinoid modulatory agents include alpha(2)-adrenoceptor modulators, peroxisome proliferator-activated receptor alpha agonists and TRPV1 antagonists. An extension of the polypharmacological approach is to combine the desired pharmacological properties of the treatment within a single molecule. Hopefully, these approaches will yield novel analgesics that do not produce the psychotropic effects that limit the medicinal use of cannabis.


Subject(s)
Analgesics/pharmacology , Cannabinoid Receptor Modulators/antagonists & inhibitors , Endocannabinoids , Pain/drug therapy , Receptor, Cannabinoid, CB2/agonists , Analgesics/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/agonists , Cannabinoid Receptor Modulators/metabolism , Cannabinoids/chemical synthesis , Cannabinoids/pharmacology , Drug Evaluation, Preclinical , Drug Therapy, Combination , Humans , Nociceptors/drug effects , Nociceptors/metabolism , Pain/metabolism , Pain/physiopathology , Receptor, Cannabinoid, CB2/metabolism , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism
12.
Neuropsychopharmacol Rep ; 40(3): 211-223, 2020 09.
Article in English | MEDLINE | ID: mdl-32543111

ABSTRACT

Alcohol use disorder (AUD) is characterized by dysfunction in motivational, mood-stress regulation, and sleep systems that interact in complex ways to heighten the risk of relapse during abstinence. Emerging data suggest that excessive and chronic alcohol use disrupts sleep homeostasis and, in abstinence, subjects with AUD are known to experience insomnia that may persist for weeks to years, which we propose to refer to as insomnia associated with alcohol cessation (IAAC). The purpose of this review is to provide an update of pharmacological approaches to therapy including compounds in development, to raise awareness of the prevalence of and unmet need in IAAC and highlight differences in treatment consideration for IAAC as compared to insomnia disorder. We performed a search of select electronic databases to identify studies of pharmacological agents used to treat sleep disturbances in abstinent or treatment-seeking patients with alcohol use disorder. The search, conducted in June 2019 and updated in December 2019, yielded 1,188 abstracts after duplicates were removed, of which 36 full-text articles were assessed for eligibility. Eighteen studies were included, 15 randomized controlled trials and three open-label studies. Several classes of medications including antidepressants, anticonvulsants, and antipsychotics have been evaluated for their effectiveness in treating sleep disturbances in abstinent or treatment-seeking patients with AUD. None of these medications are approved by the FDA for the treatment of IAAC, and the currently available evidence for these agents is limited. Randomized, controlled clinical trials are warranted to evaluate the efficacy and safety of medications in the treatment of IAAC.


Subject(s)
Alcohol Abstinence/trends , Alcoholism/complications , Alcoholism/drug therapy , Sleep Initiation and Maintenance Disorders/drug therapy , Sleep Initiation and Maintenance Disorders/etiology , Alcoholism/physiopathology , Anticonvulsants/therapeutic use , Antidepressive Agents/therapeutic use , Antipsychotic Agents/therapeutic use , Humans , Melatonin/therapeutic use , Randomized Controlled Trials as Topic/methods , Sleep Initiation and Maintenance Disorders/physiopathology
13.
World J Gastroenterol ; 23(33): 6065-6076, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28970722

ABSTRACT

AIM: To evaluate whether non-steroidal anti-inflammatory drugs (NSAIDs)-induced gastropathy is a clinically predictive model of referred visceral hypersensitivity. METHODS: Gastric ulcer pain was induced by the oral administration of indomethacin to male, CD1 mice (n = 10/group) and then assessed by measuring referred abdominal hypersensitivity to tactile application. A diverse range of pharmacological mechanisms contributing to the pain were subsequently investigated. These mechanisms included: transient receptor potential (TRP), sodium and acid-sensing ion channels (ASICs) as well as opioid receptors and guanylate cyclase C (GC-C). RESULTS: Results showed that two opioids and a GC-C agonist, morphine, asimadoline and linaclotide, respectively, the TRP antagonists, AMG9810 and HC-030031 and the sodium channel blocker, carbamazepine, elicited a dose- and/or time-dependent attenuation of referred visceral hypersensitivity, while the ASIC blocker, amiloride, was ineffective at all doses tested. CONCLUSION: Together, these findings implicate opioid receptors, GC-C, and sodium and TRP channel activation as possible mechanisms associated with visceral hypersensitivity. More importantly, these findings also validate NSAID-induced gastropathy as a sensitive and clinically predictive mouse model suitable for assessing novel molecules with potential pain-attenuating properties.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Hyperalgesia/pathology , Stomach Ulcer/complications , Visceral Pain/pathology , Acetanilides/therapeutic use , Acid Sensing Ion Channel Blockers/therapeutic use , Acid Sensing Ion Channels/metabolism , Acrylamides/therapeutic use , Amiloride/therapeutic use , Analgesics, Opioid/therapeutic use , Animals , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Humans , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Male , Mice , Morphine/therapeutic use , Pain Measurement/methods , Purines/therapeutic use , Random Allocation , Receptors, Atrial Natriuretic Factor/metabolism , Receptors, Opioid/agonists , Receptors, Opioid/metabolism , Stomach Ulcer/chemically induced , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/metabolism , Visceral Pain/etiology
14.
Bone ; 38(3): 387-93, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16203196

ABSTRACT

Changes in bone mineral density (BMD) are associated with clinical neuropathies. Following nerve injury in the rat, there is a loss of BMD, which may be related to nerve injury or reduced mechanical loading. The purpose of this study was to investigate if altered mechanical loading is solely responsible for the observed loss of BMD in neuropathic pain models. In addition, we sought to study the action of chronic bisphosphonate treatment on both neuropathy-induced osteopenia and pain. We therefore had two hypotheses: firstly, that nerve injuries can have variable effects on hind limb bone loss in rats which are not attributable to differences in the extent of hind limb disuse and, secondly, that bisphosphonate treatment can reverse bone loss in a rat mononeuropathy model, and this is not attributable to bisphosphonate effects on nociception or hind paw unweighting. Male Sprague-Dawley rats were subject to chronic constriction injury (CCI), partial sciatic nerve ligation (PSN) or L5 + L6 spinal nerve ligation (SNL). Loss of BMD, defined as a numerically lower BMD as compared to control animals, was extreme following CCI (maximum ipsilateral/contralateral difference of 0.023 +/- 0.011); BMD loss following either PSN or SNL in the rat was subtle (0.010 +/- 0.002 and 0.013 +/- 0.012 g/cm2, respectively), significant only at early time points and had resolved by 7 weeks post-surgery. Chronic bisphosphonate treatment significantly inhibited CCI-induced osteopenia in the rat without inhibiting the reduction in weight-bearing tactile allodynia or mechanical hyperalgesia. Loss of BMD is observed in rats in a variety of neuropathic pain models. Lack of correlation between neuropathy-induced bone loss and weight bearing demonstrates that the bone loss is not simply a function of reduced mechanical loading and suggests that altered bone-nerve signaling is involved. Furthermore, chronic bisphosphonate treatment inhibits neuropathy-induced osteopenia without affecting behavioral measurements of neuropathic pain. This indicates that osteopenia is not directly related to neuropathic pain behaviors.


Subject(s)
Bone Diseases, Metabolic/prevention & control , Neuralgia/physiopathology , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Alendronate/administration & dosage , Animals , Body Weight , Bone Density/drug effects , Bone Density Conservation Agents/administration & dosage , Diphosphonates/administration & dosage , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Hyperalgesia/physiopathology , Imidazoles/administration & dosage , Ligation , Male , Physical Stimulation , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Treatment Outcome , Zoledronic Acid
15.
Adv Pharmacol ; 75: 303-23, 2016.
Article in English | MEDLINE | ID: mdl-26920017

ABSTRACT

In recent years, animal behavioral models, particularly those used in pain research, have been increasingly scrutinized and criticized for their role in the poor translation of novel pharmacotherapies for chronic pain. This chapter addresses the use of animal models of pain used in drug discovery research. It highlights how, when, and why animal models of pain are used as one of the many experimental tools used to gain better understanding of target mechanisms and rank-order compounds in the iterative process of establishing structure-activity relationship. Together, these models help create an "analgesic signature" for a compound and inform the indications most likely to yield success in clinical trials. In addition, the authors discuss some often underappreciated aspects of currently used (traditional) animal models of pain, including simply applying basic pharmacological principles to study design and data interpretation as well as consideration of efficacy alongside side effect measures as part of the overall conclusion of efficacy. This is provided to add perspective regarding current efforts to develop new models and endpoints both in rodents and in larger animal species as well as assess cognitive and/or affective aspects of pain. Finally, the authors suggest ways in which efficacy evaluation in animal models of pain, whether traditional or new, might better align with clinical standards of analysis, citing examples where applying effect size and number needed to treat estimations to animal model data suggest that the efficacy bar often may be set too low preclinically to allow successful translation to the clinical setting.


Subject(s)
Chronic Pain/drug therapy , Drug Discovery , Animals , Behavior, Animal , Disease Models, Animal , Humans
16.
Pain ; 157(9): 2057-2067, 2016 09.
Article in English | MEDLINE | ID: mdl-27168361

ABSTRACT

This experimental, translational, experimental pain, single-center, randomized, double-blind, single-dose, 3-treatment, 3-period cross-over proof-of-concept volunteer trial studied the efficacy of a novel TRPV1 antagonist (V116517) on capsaicin- and UV-B-induced hyperalgesia. Heat and pressure pain thresholds, von Frey stimulus-response functions, and neurogenic inflammation were assessed together with safety. Each treatment period was 4 days. The 3 single oral treatments were 300 mg V116517, 400 mg celecoxib (a COX-2 inhibitor), and placebo. The heat pain detection and tolerance thresholds were increased significantly (P < 0.0001) by V116517. Heat pain detection and tolerance thresholds showed significantly less capsaicin hyperalgesia after V116517 (P = 0.004 and P < 0.0001, respectively). Celecoxib reduced UV-B-provoked pressure pain sensitization (P = 0.01). Laser Doppler flowmetry and erythema index after UV-B were significantly (P < 0.0001) reduced by celecoxib. Stimulus-response function in capsaicin-treated areas showed significant differences between both celecoxib and placebo and between V116517 and placebo. The body temperature showed no change, and no side effects were reported for any of the treatments. The TRPV1 antagonists and the COX-2 inhibitor showed different antihyperalgesic profiles indicating different clinical targets. In addition, the preclinical profile of V116517 in rat models of UV-B and capsaicin-induced hypersensitivity was compared with the human experimental data and overall demonstrated an alignment between 2 of the 3 end points tested. The TRPV1 antagonist showed a potent antihyperalgesic action without changing the body temperature but heat analgesia may be a potential safety issue.


Subject(s)
Pain/drug therapy , TRPV Cation Channels/antagonists & inhibitors , Adolescent , Adult , Aminopyridines/therapeutic use , Animals , Capsaicin/adverse effects , Celecoxib/therapeutic use , Cross-Over Studies , Cyclooxygenase 2 Inhibitors/therapeutic use , Disease Models, Animal , Double-Blind Method , Female , Healthy Volunteers , Humans , Male , Middle Aged , Pain/etiology , Pain Measurement , Pain Threshold/drug effects , Physical Stimulation/adverse effects , Pressure/adverse effects , Rats , Rats, Sprague-Dawley , Ultraviolet Rays/adverse effects , Young Adult
17.
Article in English | MEDLINE | ID: mdl-26589431

ABSTRACT

Whole body plethysmography using unrestrained animals is a common technique for assessing the respiratory risk of new drugs in safety pharmacology studies in rats. However, wide variations in experimental technique make cross laboratory comparison of data difficult and raise concerns that non-appropriate conditions may mask the deleterious effects of test compounds - in particular with suspected respiratory depressants. Therefore, the objective of this study was to evaluate the robustness of arterial blood gas analysis as an alternative to plethysmography in rats. We sought to do this by assessing the effect of different vehicles and times post-surgical catheterization on blood gas measurements, in addition to determining sensitivity to multiple opioids. Furthermore, we determined intra-lab variability from multiple datasets utilizing morphine and generated within a single lab and lastly, inter-lab variability was measured by comparing datasets generated in two separate labs. Overall, our data show that arterial blood gas analysis is a measure that is both flexible in terms of experimental conditions and highly sensitive to respiratory depressants, two key limitations when using plethysmography. As such, our data strongly advocate the adoption of arterial blood gas analysis as an investigative approach to reliably examine the respiratory depressant effects of opioids.


Subject(s)
Analgesics, Opioid/adverse effects , Blood Gas Analysis/standards , Respiratory Insufficiency/blood , Respiratory Insufficiency/chemically induced , Respiratory Mechanics/drug effects , Respiratory Mechanics/physiology , Animals , Blood Gas Analysis/methods , Buprenorphine/adverse effects , Dose-Response Relationship, Drug , Male , Morphine/adverse effects , Rats , Rats, Sprague-Dawley
18.
Pain ; 157(4): 901-909, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26683237

ABSTRACT

There is growing concern about lack of scientific rigor and transparent reporting across many preclinical fields of biological research. Poor experimental design and lack of transparent reporting can result in conscious or unconscious experimental bias, producing results that are not replicable. The Analgesic, Anesthetic, and Addiction Clinical Trial Translations, Innovations, Opportunities, and Networks (ACTTION) public-private partnership with the U.S. Food and Drug Administration sponsored a consensus meeting of the Preclinical Pain Research Consortium for Investigating Safety and Efficacy (PPRECISE) Working Group. International participants from universities, funding agencies, government agencies, industry, and a patient advocacy organization attended. Reduction of publication bias, increasing the ability of others to faithfully repeat experimental methods, and increased transparency of data reporting were specifically discussed. Parameters deemed essential to increase confidence in the published literature were clear, specific reporting of an a priori hypothesis and definition of primary outcome measure. Power calculations and whether measurement of minimal meaningful effect size to determine these should be a core component of the preclinical research effort provoked considerable discussion, with many but not all agreeing. Greater transparency of reporting should be driven by scientists, journal editors, reviewers, and grant funders. The conduct of high-quality science that is fully reported should not preclude novelty and innovation in preclinical pain research, and indeed, any efforts that curtail such innovation would be misguided. We believe that to achieve the goal of finding effective new treatments for patients with pain, the pain field needs to deal with these challenging issues.


Subject(s)
Analgesics/therapeutic use , Pain , Bias , Evidence-Based Medicine/methods , Humans , Pain/drug therapy , Research Design
19.
Neuropharmacology ; 48(5): 658-72, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15814101

ABSTRACT

To date, two cannabinoid receptors have been identified, CB1 and CB2. Activation of these receptors with non-selective cannabinoid receptor agonists reduces pain sensitivity in animals and humans. However, activation of CB1 receptors is also associated with central side effects, including ataxia and catalepsy. More recently, a role for selective CB2 agonists in pain modification has been demonstrated. GW405833, a selective CB2 agonist, was recently reported to partially reverse the inflammation and hyperalgesia in a rat model of acute inflammation. In the current report, we extend the characterization and therapeutic potential of this compound. For the first time, we show that GW405833 selectively binds both rat and human CB2 receptors with high affinity, where it acts as a partial agonist (approximately 50% reduction of forskolin-mediated cAMP production compared to the full cannabinoid agonist, CP55,940). We also report for the first time that intraperitoneal administration of GW405833 (0.3-100 mg/kg) to rats shows linear, dose-dependent increases in plasma levels and substantial penetration into the central nervous system. In addition, GW405833 (up to 30 mg/kg) elicits potent and efficacious antihyperalgesic effects in rodent models of neuropathic, incisional and chronic inflammatory pain, the first description of this compound in these models. In contrast, analgesia, sedation and catalepsy were not observed in this dose range, but were apparent at 100 mg/kg. Additionally, GW405833 was not antihyperalgesic against chronic inflammatory pain in CB2 knockout mice. These data support the tenet that selective CB2 receptor agonists have the potential to treat pain without eliciting the centrally-mediated side effects associated with non-selective cannabinoid agonists, and highlight the utility of GW405833 for the investigation of CB2 physiology.


Subject(s)
Anxiety/metabolism , Ataxia/metabolism , Catalepsy/metabolism , Indoles , Morpholines , Pain/metabolism , Receptor, Cannabinoid, CB2/agonists , Amines/pharmacology , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Behavior, Animal/drug effects , Benzoxazines , Binding, Competitive/drug effects , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Cyclohexanecarboxylic Acids/pharmacology , Cyclohexanols/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Gabapentin , Humans , Immunosuppressive Agents/pharmacology , Indoles/pharmacokinetics , Indoles/pharmacology , Indomethacin/pharmacology , Inflammation/drug therapy , Male , Mice , Mice, Knockout , Morpholines/pharmacokinetics , Morpholines/pharmacology , Naphthalenes/pharmacology , Pain Measurement/methods , Psychomotor Performance/drug effects , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Receptor, Cannabinoid, CB2/deficiency , Receptor, Cannabinoid, CB2/metabolism , Time Factors , gamma-Aminobutyric Acid/pharmacology
20.
Eur J Pharmacol ; 528(1-3): 65-72, 2005 Dec 28.
Article in English | MEDLINE | ID: mdl-16316650

ABSTRACT

Several recent reports have demonstrated a role for selective cannabinoid CB2 receptor agonists in pain modulation, showing both analgesic and antihyperalgesic activities. While the mechanism of action is poorly understood, it has been postulated that these effects may be indirect, involving release of endogenous opioids. We have previously reported that administration of the selective cannabinoid CB2 receptor agonist GW405833 (2,3-dichloro-phenyl)-[5-methoxy-2-methyl-3-(2-morpholin-4-yl-ethyl)-indol-1-yl]-methanone) to rats elicits potent and efficacious antihyperalgesic effects against neuropathic and inflammatory pain and, at high dose (100 mg/kg), is analgesic and ataxic [Valenzano, K.J., Tafesse, L., Lee, G., Harrison, J.E., Boulet, J., Gottshall, S.L., Mark, L., Pearson, M.S., Miller, W., Shan, S., Rabadi, L., Rotstheyn, Y., Chaffer, S.M., Turchin, P.I., Elsemore, D.A., Toth, M., Koetzner, L., Whiteside, G.T., 2005. Pharmacological and pharmacokinetic characterization of the cannabinoid receptor 2 agonist, GW405833, utilizing rodent models of acute and chronic pain, anxiety, ataxia and catalepsy. Neuropharmacology 48, 658-672]. In the current study, we confirm these properties using mouse models and investigate the role of cannabinoid CB2 receptors using knockout animals. Furthermore, we provide evidence that the antinociceptive properties of GW405833 are opioid independent. GW405833 elicited robust antihyperalgesic effects in mouse models of inflammatory (Freund's complete adjuvant) and neuropathic (Seltzer) pain. In contrast, GW405833 showed no antihyperalgesic activity against Freund's complete adjuvant-mediated inflammatory pain in cannabinoid CB2 receptor knockout mice. As in rats, high-dose GW405833 (100 mg/kg) showed both analgesic and sedative activities in wild-type mice, activities that were also apparent in cannabinoid CB2 receptor knockout mice. In rats, neither the antihyperalgesic effect in the Freund's complete adjuvant model nor the analgesic effects in tail flick and hot plate assays were inhibited by pre-treatment with the non-selective opioid receptor antagonist, naltrexone. These data demonstrate that the antihyperalgesic effects of GW405833 are mediated via the cannabinoid CB2 receptor, whereas the analgesic and sedative effects are not. Furthermore, these data suggest that the mechanism of action for GW405833 does not depend on the release of endogenous opioids.


Subject(s)
Analgesics/pharmacology , Behavior, Animal/drug effects , Indoles/pharmacology , Morpholines/pharmacology , Receptors, Cannabinoid/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Freund's Adjuvant , Hyperalgesia/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Opioid Peptides/metabolism , Pain/chemically induced , Pain/prevention & control , Pain Threshold/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/genetics , Receptors, Cannabinoid/genetics , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Sciatic Nerve/surgery
SELECTION OF CITATIONS
SEARCH DETAIL