1
|
Iglesias LP, Aguiar DC, Moreira FA. TRPV1 blockers as potential new treatments for psychiatric disorders. Behav Pharmacol 2022; 33:2-14. [PMID: 33136616 DOI: 10.1097/fbp.0000000000000603] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The transient receptor potential vanilloid-1 channel (TRPV1) is responsible for decoding physical and chemical stimuli. TRPV1 is activated by capsaicin (a compound from chili peppers), heat (above 43°C) and acid environment, playing a major role in pain, inflammation and body temperature. Molecular and histological studies have suggested TRPV1 expression in specific brain regions, where it can be activated primarily by the endocannabinoid anandamide, fostering studies on its potential role in psychiatric disorders. TRPV1 blockers are effective in various animal models predictive of anxiolytic and antipanic activities, in addition to reducing conditioned fear. In models of antidepressant activity, these compounds reduce behavioral despair and promote active stress-coping behavior. TRPV1 blockers also reduce the effects of certain drugs of abuse and revert behavioral changes in animal models of neurodevelopmental disorders. The main limiting factor in developing TRPV1 blockers as therapeutic agents concerns their effects on body temperature, particularly hyperthermia. New compounds, which block specific states of the channel, could represent an alternative. Moreover, compounds blocking both TRPV1 and the anandamide-hydrolyzing enzyme, fatty acid amide hydrolase (FAAH), termed dual TRPV1/FAAH blockers, have been investigated with promising results. Overall, preclinical studies yield favorable results with TRPV1 blockers in animal models of psychiatric disorders.
Collapse
Affiliation(s)
- Lia P Iglesias
- Department of Pharmacology, Graduate School of Neuroscience
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gera, Brazil
| | - Daniele C Aguiar
- Department of Pharmacology, Graduate School of Neuroscience
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gera, Brazil
| | - Fabrício A Moreira
- Department of Pharmacology, Graduate School of Neuroscience
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gera, Brazil
| |
Collapse
|
2
|
Bamps D, Vriens J, de Hoon J, Voets T. TRP Channel Cooperation for Nociception: Therapeutic Opportunities. Annu Rev Pharmacol Toxicol 2020; 61:655-677. [PMID: 32976736 DOI: 10.1146/annurev-pharmtox-010919-023238] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chronic pain treatment remains a sore challenge, and in our aging society, the number of patients reporting inadequate pain relief continues to grow. Current treatment options all have their drawbacks, including limited efficacy and the propensity of abuse and addiction; the latter is exemplified by the ongoing opioid crisis. Extensive research in the last few decades has focused on mechanisms underlying chronic pain states, thereby producing attractive opportunities for novel, effective and safe pharmaceutical interventions. Members of the transient receptor potential (TRP) ion channel family represent innovative targets to tackle pain sensation at the root. Three TRP channels, TRPV1, TRPM3, and TRPA1, are of particular interest, as they were identified as sensors of chemical- and heat-induced pain in nociceptor neurons. This review summarizes the knowledge regarding TRP channel-based pain therapies, including the bumpy road of the clinical development of TRPV1 antagonists, the current status of TRPA1 antagonists, and the future potential of targeting TRPM3.
Collapse
Affiliation(s)
- Dorien Bamps
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis and Reproductive Medicine, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Jan de Hoon
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research, VIB-KU Leuven Center for Brain and Disease Research, 3000 Leuven, Belgium; .,Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
3
|
Garami A, Shimansky YP, Rumbus Z, Vizin RCL, Farkas N, Hegyi J, Szakacs Z, Solymar M, Csenkey A, Chiche DA, Kapil R, Kyle DJ, Van Horn WD, Hegyi P, Romanovsky AA. Hyperthermia induced by transient receptor potential vanilloid-1 (TRPV1) antagonists in human clinical trials: Insights from mathematical modeling and meta-analysis. Pharmacol Ther 2020; 208:107474. [PMID: 31926897 DOI: 10.1016/j.pharmthera.2020.107474] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023]
Abstract
Antagonists of the transient receptor potential vanilloid-1 (TRPV1) channel alter body temperature (Tb) in laboratory animals and humans: most cause hyperthermia; some produce hypothermia; and yet others have no effect. TRPV1 can be activated by capsaicin (CAP), protons (low pH), and heat. First-generation (polymodal) TRPV1 antagonists potently block all three TRPV1 activation modes. Second-generation (mode-selective) TRPV1 antagonists potently block channel activation by CAP, but exert different effects (e.g., potentiation, no effect, or low-potency inhibition) in the proton mode, heat mode, or both. Based on our earlier studies in rats, only one mode of TRPV1 activation - by protons - is involved in thermoregulatory responses to TRPV1 antagonists. In rats, compounds that potently block, potentiate, or have no effect on proton activation cause hyperthermia, hypothermia, or no effect on Tb, respectively. A Tb response occurs when a TRPV1 antagonist blocks (in case of hyperthermia) or potentiates (hypothermia) the tonic TRPV1 activation by protons somewhere in the trunk, perhaps in muscles, and - via the acido-antithermogenic and acido-antivasoconstrictor reflexes - modulates thermogenesis and skin vasoconstriction. In this work, we used a mathematical model to analyze Tb data from human clinical trials of TRPV1 antagonists. The analysis suggests that, in humans, the hyperthermic effect depends on the antagonist's potency to block TRPV1 activation not only by protons, but also by heat, while the CAP activation mode is uninvolved. Whereas in rats TRPV1 drives thermoeffectors by mediating pH signals from the trunk, but not Tb signals, our analysis suggests that TRPV1 mediates both pH and thermal signals driving thermoregulation in humans. Hence, in humans (but not in rats), TRPV1 is likely to serve as a thermosensor of the thermoregulation system. We also conducted a meta-analysis of Tb data from human trials and found that polymodal TRPV1 antagonists (ABT-102, AZD1386, and V116517) increase Tb, whereas the mode-selective blocker NEO6860 does not. Several strategies of harnessing the thermoregulatory effects of TRPV1 antagonists in humans are discussed.
Collapse
Affiliation(s)
- Andras Garami
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary.
| | - Yury P Shimansky
- Department of Neurobiology, Barrow Neurological Institute, Dignity Health, Phoenix, AZ, USA
| | - Zoltan Rumbus
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Robson C L Vizin
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), Trauma Research, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, AZ, USA
| | - Nelli Farkas
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Judit Hegyi
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Zsolt Szakacs
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Margit Solymar
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Alexandra Csenkey
- Department of Thermophysiology, Institute for Translational Medicine, Medical School, University of Pecs, Pecs, Hungary
| | | | | | | | - Wade D Van Horn
- School of Molecular Sciences, Arizona State University, Tempe, AZ, USA
| | - Peter Hegyi
- Institute for Translational Medicine, Medical School and Szentagothai Research Centre, University of Pecs, Pecs, Hungary; Department of Translational Medicine, First Department of Medicine, Medical School, University of Pecs, Pecs, Hungary
| | - Andrej A Romanovsky
- Thermoregulation and Systemic Inflammation Laboratory (FeverLab), Trauma Research, St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, AZ, USA; School of Molecular Sciences, Arizona State University, Tempe, AZ, USA; Zharko Pharma Inc., Olympia, WA, USA.
| |
Collapse
|
4
|
Schaffler K, He W, Passier P, Tracy K, Fakhoury A, Paul J. A Phase I, Randomized, Double-Blind, Laser-Evoked Potential Study to Evaluate the Analgesic/Antihyperalgesic Effect of ASP9226, a State-Dependent N-Type Voltage-Gated Calcium Channel Inhibitor, in Healthy Male Subjects. PAIN MEDICINE 2019; 19:2246-2255. [PMID: 29378016 PMCID: PMC6217710 DOI: 10.1093/pm/pnx338] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Objective Evaluate the analgesic/antihyperalgesic effects of ASP9226, a state-dependent N-type voltage-gated calcium channel inhibitor, in healthy male subjects. Design Randomized, double-blind, double-dummy, placebo- and active comparator–controlled crossover study. Setting HPR Dr. Schaffler GmbH, Munich, Germany. Subject Healthy male subjects aged 18–55 years. Methods Twenty-four eligible subjects were randomly assigned to one of four treatment sequences and received single doses of ASP9226 (30 mg or 50 mg), pregabalin (150 mg), or placebo during four treatment periods. Laser-evoked potentials (LEP) and postlaser pain visual analog scales (VAS) on capsaicin-treated skin were assessed during main assessment days (the first day of each study period). Primary and secondary end points were the differences in LEP N2-P2 peak-to-peak (PtP) amplitudes and VAS score, respectively, in all subjects. Results Overall, treatment with pregabalin resulted in a significantly lower LEP N2-P2 PtP amplitude vs placebo (–3.30 μV, P < 0.0001). There were no clinically relevant differences in N2-P2 PtP amplitudes between placebo and either ASP9226 dose (–0.31 μV and –0.27 μV). Furthermore, subjects reported significantly lower VAS pain scores with pregabalin vs placebo (–9.90%, P < 0.0001) in contrast to ASP9226 30 mg (–2.1%) and ASP9226 50 mg (1.2%) vs placebo. Subgroup analysis of LEP and VAS pain in participants with positive prestudy capsaicin response (n = 13) were in keeping with results in all subjects. Conclusions ASP9226 was well tolerated; however, there was no improvement in LEP and VAS pain scores with ASP9226 at either dose vs placebo.
Collapse
Affiliation(s)
- Klaus Schaffler
- HPR (Human Pharmacodynamic Research) Dr. Schaffler GmbH, Munich, Germany
| | - Weizhong He
- Astellas Pharma Global Development, Northbrook, Illinois, USA
| | - Paul Passier
- Astellas Pharma Europe B.V., Leiden, the Netherlands
| | - Katherine Tracy
- Astellas Pharma Global Development, Northbrook, Illinois, USA
| | - Allam Fakhoury
- Astellas Pharma Global Development, Northbrook, Illinois, USA
| | - Jeffrey Paul
- Astellas Pharma Global Development, Northbrook, Illinois, USA
| |
Collapse
|
5
|
Schaffler K, Yassen A, Reeh P, Passier P. A Randomized, Double-Blind, Placebo- and Active Comparator-Controlled Phase I Study of Analgesic/Antihyperalgesic Properties of ASP8477, a Fatty Acid Amide Hydrolase Inhibitor, in Healthy Female Subjects. PAIN MEDICINE 2019; 19:1206-1218. [PMID: 29228247 PMCID: PMC5998989 DOI: 10.1093/pm/pnx281] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objectives To evaluate the analgesic/antihyperalgesic effect of ASP8477. Design Randomized, double-blind, double-dummy, cross-over, placebo- and active comparator-controlled study. Setting HPR Dr. Schaffler GmbH, Munich, Germany. Subjects Healthy female subjects aged 18–65 years. Methods Eligible subjects were randomly assigned to one of six treatment sequences and received multiple ascending doses of ASP8477, duloxetine, and placebo over three treatment periods (each consisting of 21-day dosing separated by 14-day washout periods). On the last day of each dose level, laser evoked potentials (LEPs) and visual analog scales (VAS pain) on capsaicin-treated skin at baseline and at multiple postdose time points were assessed. The primary end point was the difference in LEP N2-P2 peak-to-peak (PtP) amplitudes for ASP8477 100 mg vs placebo. Results Twenty-five subjects were randomized. In all subjects, LEP N2-P2 PtP amplitudes were numerically lower for ASP8477 100 mg vs placebo (P = 0.0721); in subjects who demonstrated positive capsaicin skin effects, a greater mean difference of –2.24 µV (P = 0.0146) was observed. Across all doses, LEP N2-P2 PtP amplitudes were lower for duloxetine compared with ASP8477 (mean difference –3.80 µV; P < 0.0001) or placebo (mean difference –5.21 µV; P < 0.0001). The effect of ASP8477 (all doses) on down-scoring the VAS pain score was significant compared with placebo (mean difference –2.55%; P < 0.0007). Conclusions ASP8477 was well tolerated in this study. Analysis of all subjects did not demonstrate a significant difference in LEP for ASP8477 100 mg over placebo but did in subjects who demonstrated positive capsaicin skin effects.
Collapse
Affiliation(s)
- Klaus Schaffler
- Human Pharmacodynamic Research (HPR) Dr. Schaffler GmbH, Munich, Germany
| | | | - Peter Reeh
- Institute for Physiology and Pathophysiology, University Erlangen-Nuremberg, Erlangen, Germany
| | - Paul Passier
- Astellas Pharma Europe B.V., Leiden, Netherlands
| |
Collapse
|
6
|
Quantitative sensory testing response patterns to capsaicin- and ultraviolet-B-induced local skin hypersensitization in healthy subjects: a machine-learned analysis. Pain 2019; 159:11-24. [PMID: 28700537 PMCID: PMC5737455 DOI: 10.1097/j.pain.0000000000001008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The comprehensive assessment of pain-related human phenotypes requires combinations of nociceptive measures that produce complex high-dimensional data, posing challenges to bioinformatic analysis. In this study, we assessed established experimental models of heat hyperalgesia of the skin, consisting of local ultraviolet-B (UV-B) irradiation or capsaicin application, in 82 healthy subjects using a variety of noxious stimuli. We extended the original heat stimulation by applying cold and mechanical stimuli and assessing the hypersensitization effects with a clinically established quantitative sensory testing (QST) battery (German Research Network on Neuropathic Pain). This study provided a 246 × 10-sized data matrix (82 subjects assessed at baseline, following UV-B application, and following capsaicin application) with respect to 10 QST parameters, which we analyzed using machine-learning techniques. We observed statistically significant effects of the hypersensitization treatments in 9 different QST parameters. Supervised machine-learned analysis implemented as random forests followed by ABC analysis pointed to heat pain thresholds as the most relevantly affected QST parameter. However, decision tree analysis indicated that UV-B additionally modulated sensitivity to cold. Unsupervised machine-learning techniques, implemented as emergent self-organizing maps, hinted at subgroups responding to topical application of capsaicin. The distinction among subgroups was based on sensitivity to pressure pain, which could be attributed to sex differences, with women being more sensitive than men. Thus, while UV-B and capsaicin share a major component of heat pain sensitization, they differ in their effects on QST parameter patterns in healthy subjects, suggesting a lack of redundancy between these models.
Collapse
|
7
|
Mayorga AJ, Flores CM, Trudeau JJ, Moyer JA, Shalayda K, Dale M, Frustaci ME, Katz N, Manitpisitkul P, Treister R, Ratcliffe S, Romano G. A randomized study to evaluate the analgesic efficacy of a single dose of the TRPV1 antagonist mavatrep in patients with osteoarthritis. Scand J Pain 2017; 17:134-143. [PMID: 28850367 DOI: 10.1016/j.sjpain.2017.07.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 07/13/2017] [Accepted: 07/25/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND/AIMS Transient receptor potential vanilloid type 1 (TRPV1) receptor antagonists have been evaluated in clinical studies for their analgesic effects. Mavatrep, a potent, selective, competitive TRPV1 receptor antagonist has demonstrated pharmacodynamic effects consistent with target engagement at the TRPV1 receptor in a previous single-dose clinical study. The current study was conducted to evaluate the analgesic effects of a single dose of mavatrep. METHODS In this randomized, placebo- and active-controlled, 3-way crossover, phase 1b study, patients with painful knee osteoarthritis were treated with a single-dose of 50mg mavatrep, 500mg naproxen twice-daily, and placebo. Patients were randomized to 1 of 6 treatment sequences. Each treatment sequence included three treatment periods of 7 days duration with a 7 day washout between each treatment period. The primary efficacy evaluation was pain reduction measured by the 4-h postdose sum of pain intensity difference (SPID) based on the 11-point (0-10) Numerical Rating Scale (NRS) for pain after stair-climbing (PASC). The secondary efficacy evaluations included 11-point (0-10) NRS pain scores entered into the Actiwatch between clinic visits, the Western Ontario and McMaster Universities Arthritis Index subscales (WOMAC) questionnaire, and use of rescue medication. Safety and tolerability of single oral dose mavatrep were also assessed. RESULTS Of 33 patients randomized, 32 completed the study. A statistically significantly (p<0.1) greater reduction in PASC was observed for mavatrep versus placebo (4-h SPID least square mean [LSM] [SE] difference: 1.5 [0.53]; p=0.005 and 2-h LSM [SE] difference of PID: 0.7 [0.30]; p=0.029). The mean average daily current pain NRS scores were lower in the mavatrep and naproxen treatment arm than in the placebo arm (mavatrep: 7 day mean [SD], 3.72 [1.851]; naproxen: 7 day mean [SD], 3.49 [1.544]; placebo: 7 day mean [SD], 4.9 [1.413]). Mavatrep showed statistically significant improvements as compared with placebo on the WOMAC subscales (pain on days 2 [p=0.049] and 7 [p=0.041], stiffness on day 7 [p=0.075]), and function on day 7 [p=0.077]). The same pattern of improvement was evident for naproxen versus placebo. The mean (SD) number of rescue medication tablets taken during the 7-day treatment period was 4.2 (6.49) for mavatrep treatment, 2.8 (5.42) for naproxen, and 6.3 (8.25) for placebo treatment. All patients that received mavatrep reported at least 1 treatment emergent adverse event (TEAE). Feeling cold (79%), thermohypoesthesia (61%), dysgeusia (58%), paraesthesia (36%), and feeling hot (15%) were the most common TEAEs in the mavatrep group. Total 9% patients receiving mavatrep experienced minor thermal burns. No deaths or serious AEs or discontinuations due to AEs occurred. CONCLUSION Overall, mavatrep was associated with a significant reduction in pain, stiffness, and physical function when compared with placebo in patients with knee osteoarthritis. Mavatrep's safety profile was consistent with its mechanism of action as a TRPV1 antagonist. IMPLICATIONS Further studies are required to evaluate whether lower multiple doses of mavatrep can produce analgesic efficacy while minimizing adverse events, as well as the potential for improved patient counselling techniques to reduce the minor thermal burns related to decreased heat perception. TRIAL REGISTRATION 2009-010961-21 (EudraCT Number).
Collapse
Affiliation(s)
| | | | | | - John A Moyer
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Kevin Shalayda
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Mark Dale
- MAC Clinical Research, Manchester, UK
| | | | - Nathaniel Katz
- Analgesic Solutions, Natick, MA, USA; Tufts University School of Medicine, Boston, MA, USA
| | | | - Roi Treister
- Analgesic Solutions, Natick, MA, USA; Tufts University School of Medicine, Boston, MA, USA; Massachusetts General Hospital & Harvard Medical School, Department of Neurology, Nerve Injury Unit, Boston, MA, USA
| | | | - Gary Romano
- Janssen Research & Development, LLC, Titusville, NJ, USA
| |
Collapse
|
8
|
Schaffler K, Nicolas LB, Borta A, Brand T, Reitmeir P, Roebling R, Scholpp J. Investigation of the predictive validity of laser-EPs in normal, UVB-inflamed and capsaicin-irritated skin with four analgesic compounds in healthy volunteers. Br J Clin Pharmacol 2017; 83:1424-1435. [PMID: 28139023 DOI: 10.1111/bcp.13247] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 01/24/2017] [Accepted: 01/27/2017] [Indexed: 11/28/2022] Open
Abstract
AIMS The aim of the present study was to assess the predictivity of laser-(radiant-heat)-evoked potentials (LEPs) from the vertex electroencephalogram, using an algesimetric procedure, testing the anti-nociceptive/anti-hyperalgesic effects of single oral doses of four marketed analgesics (of different compound classes) vs. placebo, in healthy volunteers with three skin types. METHODS This was a randomized, placebo-controlled, single-blind, five-way-crossover trial. Twenty-five healthy male/female Caucasians were included (receiving celecoxib 200 mg, pregabalin 150 mg, duloxetine 60 mg, lacosamide 100 mg or placebo) in a Williams design, with CO2 laser-induced painful stimuli to normal, ultraviolet (UV) B-inflamed and capsaicin-irritated skin. LEPs and visual analogue scale ratings were taken at baseline and hourly for 6 h postdose from all three skin types. RESULTS In normal skin, the averaged postdose LEP peak-to-peak-(PtP)-amplitudes were reduced by pregabalin (-2.68 μV; 95% confidence interval (CI) -4.16, 1.19) and duloxetine (-1.73 μV; 95% CI -3.21, -0.26) but not by lacosamide and celecoxib vs. placebo. On UVB-irradiated skin, reflecting inflammatory pain, celecoxib induced a pronounced reduction in LEP PtP amplitudes vs. placebo (-6.2 μV; 95% CI -7.88, -4.51), with a smaller reduction by duloxetine (-4.54 μV; 95% CI -6.21, -2.87) and pregabalin (-3.72 μV; 95% CI -5.40, -2.04), whereas lacosamide was inactive. LEP PtP amplitudes on capsaicin-irritated skin, reflecting peripheral/spinal sensitization, as in neuropathic pain, were reduced by pregabalin (-3.78 μV; 95% CI -5.31, -2.25) and duloxetine (-2.32 μV; 95% CI -3.82, -0.82) but not by celecoxib or lacosamide vs. placebo, which was in agreement with known clinical profiles. Overall, PtP amplitude reductions were in agreement with subjective ratings. CONCLUSIONS LEP algesimetry is sensitive to analgesics with different modes of action and may enable the effects of novel analgesics to be assessed during early clinical development.
Collapse
Affiliation(s)
| | - Laurent B Nicolas
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Andreas Borta
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Tobias Brand
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Peter Reitmeir
- HPR, Human Pharmacodynamic Research GmbH, Munich, Germany
| | - Robert Roebling
- Medicine, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Joachim Scholpp
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
9
|
Buntinx L, Voets T, Morlion B, Vangeel L, Janssen M, Cornelissen E, Vriens J, de Hoon J, Levtchenko E. TRPV1 dysfunction in cystinosis patients harboring the homozygous 57 kb deletion. Sci Rep 2016; 6:35395. [PMID: 27734949 PMCID: PMC5062165 DOI: 10.1038/srep35395] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/27/2016] [Indexed: 11/12/2022] Open
Abstract
Cystinosis is a rare autosomal recessive disorder characterized by lysosomal cystine accumulation due to loss of function of the lysosomal cystine transporter (CTNS). The most common mutation in cystinosis patients of Northern Europe consists of a 57-kb deletion. This deletion not only inactivates the CTNS gene but also extends into the non-coding region upstream of the start codon of the TRPV1 gene, encoding the capsaicin- and heat-sensitive ion channel TRPV1. To evaluate the consequences of the 57-kb deletion on functional TRPV1 expression, we compared thermal, mechanical and chemical sensitivity of cystinosis patients with matched healthy controls. Whereas patients heterozygous for the 57-kb deletion showed normal sensory responses, homozygous subjects exhibited a 60% reduction in vasodilation and pain evoked by capsaicin, as well as an increase in heat detection threshold. Responses to cold, mechanical stimuli or cinnamaldehyde, an agonist of the related nociceptor channel TRPA1, were unaltered. We conclude that cystinosis patients homozygous for the 57-kb deletion exhibit a strong reduction of TRPV1 function, leading to sensory deficiencies akin to the phenotype of TRPV1-deficient mice. These deficits may account for the reported sensory alterations and thermoregulatory deficits in these patients, and provide a paradigm for life-long TRPV1 deficiency in humans.
Collapse
Affiliation(s)
- L Buntinx
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - T Voets
- Department of Cellular and Molecular Medicine, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - B Morlion
- Center for algology and pain management, Department of Cardiovascular Sciences, KULeuven, Weligerveld 1, 3212 Pellenberg, Belgium
| | - L Vangeel
- Department of Cellular and Molecular Medicine, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - M Janssen
- Department of internal medicine, Radboud UMC Nijmegen, Geert Grooteplein-Zuid 22, 6525 GA Nijmegen, The Netherlands
| | - E Cornelissen
- Department of Pediatric Nephrology, Radboud UMC Nijmegen, Geert Grooteplein-Zuid 22, 6525 GA Nijmegen, The Netherlands
| | - J Vriens
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - J de Hoon
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - E Levtchenko
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
10
|
A randomized, double-blind, positive-controlled, 3-way cross-over human experimental pain study of a TRPV1 antagonist (V116517) in healthy volunteers and comparison with preclinical profile. Pain 2016; 157:2057-2067. [DOI: 10.1097/j.pain.0000000000000610] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
11
|
Polymodal Transient Receptor Potential Vanilloid Type 1 Nocisensor. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 104:81-125. [DOI: 10.1016/bs.apcsb.2015.11.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
12
|
Yan L, Wang J, Pan M, Qiu Q, Huang W, Qian H. Synthesis of Analogues of BCTC Incorporating a Pyrrolidinyl Linker and Biological Evaluation as Transient Receptor Potential Vanilloid 1 Antagonists. Chem Biol Drug Des 2015; 87:306-11. [DOI: 10.1111/cbdd.12661] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 08/12/2015] [Accepted: 09/06/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Lin Yan
- Institute of Chemistry & Biology; Henan University; Kaifeng 475004 China
| | - Jingjie Wang
- State Key Laboratory of Natural Medicines; Center of Drug Discovery; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 China
| | - Miaobo Pan
- State Key Laboratory of Natural Medicines; Center of Drug Discovery; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 China
| | - Qianqian Qiu
- State Key Laboratory of Natural Medicines; Center of Drug Discovery; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 China
| | - Wenlong Huang
- State Key Laboratory of Natural Medicines; Center of Drug Discovery; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 China
| | - Hai Qian
- State Key Laboratory of Natural Medicines; Center of Drug Discovery; China Pharmaceutical University; 24 Tongjiaxiang Nanjing 210009 China
| |
Collapse
|
13
|
Lopes DM, McMahon SB. Ultraviolet Radiation on the Skin: A Painful Experience? CNS Neurosci Ther 2015; 22:118-26. [PMID: 26331607 PMCID: PMC4833175 DOI: 10.1111/cns.12444] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 07/21/2015] [Accepted: 07/22/2015] [Indexed: 12/31/2022] Open
Abstract
Excessive exposure of skin to ultraviolet radiation (UVR) has dramatic clinical effects in humans, and it is a significant public health concern. Discomfort and sensory changes caused by skin sunburn are the main common features experienced by many of us, a phenomena triggered by the combination of long and short wavelengths radiation (UVA and UVB, respectively). Although the biological processes underlying UVR exposure are not fully understood, in the last few years many studies have made significant progress in characterizing sunburn at the cellular and molecular levels, making use of both humans and laboratory animal models. Here we review and reason that UVR can be used as an excellent model of sensitization and inflammation for pain research. UVR, particularly UVB, produces a controllable and sterile inflammation that causes a robust dose‐dependent hypersensitivity with minimal confounding effects. Importantly, we show that UVR animal models precisely recapitulate the sensory, cellular, and molecular changes observed in human skin, giving it great confidence as a translational model. Furthermore, in this article, we give an overview of the pharmacology underlying UVB inflammation, the latest advances in the field, and potential new targets for inflammatory pain.
Collapse
Affiliation(s)
- Douglas M Lopes
- Neurorestoration group, Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Stephen B McMahon
- Neurorestoration group, Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| |
Collapse
|
14
|
Borsook D, Hargreaves R, Bountra C, Porreca F. Lost but making progress--Where will new analgesic drugs come from? Sci Transl Med 2015; 6:249sr3. [PMID: 25122640 DOI: 10.1126/scitranslmed.3008320] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There is a critical need for effective new pharmacotherapies for pain. The paucity of new drugs successfully reaching the clinic calls for a reassessment of current analgesic drug discovery approaches. Many points early in the discovery process present significant hurdles, making it critical to exploit advances in pain neurobiology to increase the probability of success. In this review, we highlight approaches that are being pursued vigorously by the pain community for drug discovery, including innovative preclinical pain models, insights from genetics, mechanistic phenotyping of pain patients, development of biomarkers, and emerging insights into chronic pain as a disorder of both the periphery and the brain. Collaborative efforts between pharmaceutical, academic, and public entities to advance research in these areas promise to de-risk potential targets, stimulate investment, and speed evaluation and development of better pain therapies.
Collapse
Affiliation(s)
- David Borsook
- Center for Pain and the Brain, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Richard Hargreaves
- Center for Pain and the Brain, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Chas Bountra
- Department of Clinical Medicine, University of Oxford, Oxford OX1 2JD, UK
| | - Frank Porreca
- Center for Pain and the Brain and Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA.
| |
Collapse
|
15
|
Nash MS, Verkuyl JM, Bhalay G. TRPV1 Antagonism: From Research to Clinic. ION CHANNEL DRUG DISCOVERY 2014. [DOI: 10.1039/9781849735087-00186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The capsaicin receptor, TRPV1, has been one of the most extensively studied molecules in sensory research. Its contribution to the sensation of pain in numerous pre-clinical inflammatory and neuropathic paradigms has been well-established and expression analysis suggests a potential role clinically in pain and bladder conditions. The field has now reached an exciting point in time with the development of a number of high quality TRPV1 antagonist drug candidates and the release of clinical data. What has become apparent from this work is that inhibition of TRPV1 function brings with it the potential liabilities of increased body temperature and altered thermal perception. However, there is cause for optimism because it appears that not all antagonists have the same properties and compounds can be identified that lack significant on-target side-effects whilst retaining efficacy, at least pre-clinically. What is perhaps now more critical to address is the question of how effective the analgesia provided by a TRPV1 antagonist will be. Although tantalizing clinical data showing effects on experimentally-induced pain or pain following molar extraction have been reported, no clear efficacy in a chronic pain condition has yet been demonstrated making it difficult to perform an accurate risk-benefit analysis for TRPV1 antagonists. Here we provide an overview of some of the most advanced clinical candidates and discuss the approaches being taken to avoid the now well established on-target effects of TRPV1 antagonists.
Collapse
Affiliation(s)
- Mark S. Nash
- Novartis Institutes for Biomedical Research Forum 1, Novartis Campus CH - 4056 Basel Switzerland
| | - J. Martin Verkuyl
- Novartis Institutes for Biomedical Research Wimblehurst Road Horsham, West Sussex RH12 5AB UK
| | - Gurdip Bhalay
- Novartis Institutes for Biomedical Research Wimblehurst Road Horsham, West Sussex RH12 5AB UK
| |
Collapse
|
16
|
Walters ET. Neuroinflammatory contributions to pain after SCI: roles for central glial mechanisms and nociceptor-mediated host defense. Exp Neurol 2014; 258:48-61. [PMID: 25017887 DOI: 10.1016/j.expneurol.2014.02.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/23/2014] [Accepted: 02/02/2014] [Indexed: 12/30/2022]
Abstract
Neuropathic pain after spinal cord injury (SCI) is common, often intractable, and can be severely debilitating. A number of mechanisms have been proposed for this pain, which are discussed briefly, along with methods for revealing SCI pain in animal models, such as the recently applied conditioned place preference test. During the last decade, studies of animal models have shown that both central neuroinflammation and behavioral hypersensitivity (indirect reflex measures of pain) persist chronically after SCI. Interventions that reduce neuroinflammation have been found to ameliorate pain-related behavior, such as treatment with agents that inhibit the activation states of microglia and/or astroglia (including IL-10, minocycline, etanercept, propentofylline, ibudilast, licofelone, SP600125, carbenoxolone). Reversal of pain-related behavior has also been shown with disruption by an inhibitor (CR8) and/or genetic deletion of cell cycle-related proteins, deletion of a truncated receptor (trkB.T1) for brain-derived neurotrophic factor (BDNF), or reduction by antisense knockdown or an inhibitor (AMG9810) of the activity of channels (TRPV1 or Nav1.8) important for electrical activity in primary nociceptors. Nociceptor activity is known to drive central neuroinflammation in peripheral injury models, and nociceptors appear to be an integral component of host defense. Thus, emerging results suggest that spinal and systemic effects of SCI can activate nociceptor-mediated host defense responses that interact via neuroinflammatory signaling with complex central consequences of SCI to drive chronic pain. This broader view of SCI-induced neuroinflammation suggests new targets, and additional complications, for efforts to develop effective treatments for neuropathic SCI pain.
Collapse
Affiliation(s)
- Edgar T Walters
- Department of Integrative Biology and Pharmacology, University of Texas Medical School at Houston, TX, USA.
| |
Collapse
|
17
|
Tafesse L, Kanemasa T, Kurose N, Yu J, Asaki T, Wu G, Iwamoto Y, Yamaguchi Y, Ni C, Engel J, Tsuno N, Patel A, Zhou X, Shintani T, Brown K, Hasegawa T, Shet M, Iso Y, Kato A, Kyle DJ. Structure–Activity Relationship Studies and Discovery of a Potent Transient Receptor Potential Vanilloid (TRPV1) Antagonist 4-[3-Chloro-5-[(1S)-1,2-dihydroxyethyl]-2-pyridyl]-N-[5-(trifluoromethyl)-2-pyridyl]-3,6-dihydro-2H-pyridine-1-carboxamide (V116517) as a Clinical Candidate for Pain Management. J Med Chem 2014; 57:6781-94. [DOI: 10.1021/jm500818a] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Laykea Tafesse
- Discovery
Research, Purdue Pharma LP, 6 Cedar Brook Drive, Cranbury, New Jersey 08512, United States
| | - Toshiyuki Kanemasa
- Shionogi Research Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-chow, Toyonaka, Osaka, Japan
| | - Noriyuki Kurose
- Shionogi Research Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-chow, Toyonaka, Osaka, Japan
| | - Jianming Yu
- Discovery
Research, Purdue Pharma LP, 6 Cedar Brook Drive, Cranbury, New Jersey 08512, United States
| | - Toshiyuki Asaki
- Shionogi Research Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-chow, Toyonaka, Osaka, Japan
| | - Gang Wu
- Discovery
Research, Purdue Pharma LP, 6 Cedar Brook Drive, Cranbury, New Jersey 08512, United States
| | - Yuka Iwamoto
- Shionogi Research Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-chow, Toyonaka, Osaka, Japan
| | - Yoshitaka Yamaguchi
- Shionogi Research Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-chow, Toyonaka, Osaka, Japan
| | - Chiyou Ni
- Discovery
Research, Purdue Pharma LP, 6 Cedar Brook Drive, Cranbury, New Jersey 08512, United States
| | - John Engel
- Discovery
Research, Purdue Pharma LP, 6 Cedar Brook Drive, Cranbury, New Jersey 08512, United States
| | - Naoki Tsuno
- Shionogi Research Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-chow, Toyonaka, Osaka, Japan
| | - Aniket Patel
- Discovery
Research, Purdue Pharma LP, 6 Cedar Brook Drive, Cranbury, New Jersey 08512, United States
| | - Xiaoming Zhou
- Discovery
Research, Purdue Pharma LP, 6 Cedar Brook Drive, Cranbury, New Jersey 08512, United States
| | - Takuya Shintani
- Shionogi Research Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-chow, Toyonaka, Osaka, Japan
| | - Kevin Brown
- Discovery
Research, Purdue Pharma LP, 6 Cedar Brook Drive, Cranbury, New Jersey 08512, United States
| | - Tsuyoshi Hasegawa
- Shionogi Research Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-chow, Toyonaka, Osaka, Japan
| | - Manjunath Shet
- Discovery
Research, Purdue Pharma LP, 6 Cedar Brook Drive, Cranbury, New Jersey 08512, United States
| | - Yasuyoshi Iso
- Shionogi Research Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-chow, Toyonaka, Osaka, Japan
| | - Akira Kato
- Shionogi Research Laboratories, Shionogi & Co., Ltd., 3-1-1 Futaba-chow, Toyonaka, Osaka, Japan
| | - Donald J. Kyle
- Discovery
Research, Purdue Pharma LP, 6 Cedar Brook Drive, Cranbury, New Jersey 08512, United States
| |
Collapse
|
18
|
Vriens J, Nilius B, Voets T. Peripheral thermosensation in mammals. Nat Rev Neurosci 2014; 15:573-89. [PMID: 25053448 DOI: 10.1038/nrn3784] [Citation(s) in RCA: 256] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Our ability to perceive temperature is crucial: it enables us to swiftly react to noxiously cold or hot objects and helps us to maintain a constant body temperature. Sensory nerve endings, upon depolarization by temperature-gated ion channels, convey electrical signals from the periphery to the CNS, eliciting a sense of temperature. In the past two decades, we have witnessed important advances in our understanding of mammalian thermosensation, with the identification and animal-model assessment of candidate molecular thermosensors - such as types of transient receptor potential (TRP) cation channels - involved in peripheral thermosensation. Ongoing research aims to understand how these miniature thermometers operate at the cellular and molecular level, and how they can be pharmacologically targeted to treat pain without disturbing vital thermoregulatory processes.
Collapse
Affiliation(s)
- Joris Vriens
- Laboratory of Experimental Gynaecology, KU Leuven, Herestraat 49 BOX 611, B-3000 Leuven, Belgium
| | - Bernd Nilius
- Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe), KU Leuven, Herestraat 49 BOX 802, B-3000 Leuven, Belgium
| | - Thomas Voets
- Laboratory of Ion Channel Research and TRP Research Platform Leuven (TRPLe), KU Leuven, Herestraat 49 BOX 802, B-3000 Leuven, Belgium
| |
Collapse
|
19
|
Stich AN, Rosenkrantz WS, Griffin CE. Clinical efficacy of low-level laser therapy on localized canine atopic dermatitis severity score and localized pruritic visual analog score in pedal pruritus due to canine atopic dermatitis. Vet Dermatol 2014; 25:464-e74. [PMID: 24909192 DOI: 10.1111/vde.12144] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Canine atopic dermatitis is a genetically predisposed inflammatory skin disease often requiring multimodal treatment. There is a need to find further low-risk adjunctive therapies. HYPOTHESIS/OBJECTIVES To evaluate the localized effect of low-level laser therapy (LLLT) on the paws of dogs with atopic dermatitis using a localized canine atopic dermatitis severity score (LCADSS) and owner localized pruritic visual analog score (LPVAS) in comparison to treatment with a placebo. ANIMALS Thirty client-owned dogs with symmetrical pedal pruritus due to canine atopic dermatitis. METHODS Dogs were randomly assigned into two groups. In each group, one paw was treated with LLLT and one paw treated with a placebo laser (comparing either both fore- or hindpaws). Treatments were administered at 4 J/cm(2) (area from carpus/tarsus to distal aspect of digit 3) three times per week for the first 2 weeks and two times per week for the second 2 weeks. Scores were assessed for each paw at weeks 0, 2, 4 and 5. RESULTS There were no significant differences in LCADSS or LPVAS between LLLT and placebo treatments between weeks 0 and 5 (P = 0.0856 and 0.5017, respectively). However, LCADSS and LPVAS significantly decreased from week 0 at weeks 2, 4 and 5 in both LLLT and placebo groups (P < 0.0001 for all). CONCLUSIONS AND CLINICAL IMPORTANCE Low-level laser therapy is not an effective localized treatment for pedal pruritus in canine atopic dermatitis.
Collapse
Affiliation(s)
- Ashley N Stich
- Animal Dermatology Clinic, 2965 Edinger Avenue, Tustin, CA, 92780, USA
| | | | | |
Collapse
|
20
|
Othman AA, Nothaft W, Awni WM, Dutta S. Effects of the TRPV1 antagonist ABT-102 on body temperature in healthy volunteers: pharmacokinetic/ pharmacodynamic analysis of three phase 1 trials. Br J Clin Pharmacol 2013; 75:1029-40. [PMID: 22966986 DOI: 10.1111/j.1365-2125.2012.04405.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/23/2012] [Indexed: 11/28/2022] Open
Abstract
AIM To characterize quantitatively the relationship between ABT-102, a potent and selective TRPV1 antagonist, exposure and its effects on body temperature in humans using a population pharmacokinetic/pharmacodynamic modelling approach. METHODS Serial pharmacokinetic and body temperature (oral or core) measurements from three double-blind, randomized, placebo-controlled studies [single dose (2, 6, 18, 30 and 40 mg, solution formulation), multiple dose (2, 4 and 8 mg twice daily for 7 days, solution formulation) and multiple-dose (1, 2 and 4 mg twice daily for 7 days, solid dispersion formulation)] were analyzed. NONMEM was used for model development and the model building steps were guided by pre-specified diagnostic and statistical criteria. The final model was qualified using non-parametric bootstrap and visual predictive check. RESULTS The developed body temperature model included additive components of baseline, circadian rhythm (cosine function of time) and ABT-102 effect (Emax function of plasma concentration) with tolerance development (decrease in ABT-102 Emax over time). Type of body temperature measurement (oral vs. core) was included as a fixed effect on baseline, amplitude of circadian rhythm and residual error. The model estimates (95% bootstrap confidence interval) were: baseline oral body temperature, 36.3 (36.3, 36.4)°C; baseline core body temperature, 37.0 (37.0, 37.1)°C; oral circadian amplitude, 0.25 (0.22, 0.28)°C; core circadian amplitude, 0.31 (0.28, 0.34)°C; circadian phase shift, 7.6 (7.3, 7.9) h; ABT-102 Emax , 2.2 (1.9, 2.7)°C; ABT-102 EC50 , 20 (15, 28) ng ml(-1) ; tolerance T50 , 28 (20, 43) h. CONCLUSIONS At exposures predicted to exert analgesic activity in humans, the effect of ABT-102 on body temperature is estimated to be 0.6 to 0.8°C. This effect attenuates within 2 to 3 days of dosing.
Collapse
Affiliation(s)
- Ahmed A Othman
- Abbott Clinical Pharmacology & Pharmacometrics, Abbott Laboratories, Abbott Park, IL 60064, USA.
| | | | | | | |
Collapse
|
21
|
UVB radiation generates sunburn pain and affects skin by activating epidermal TRPV4 ion channels and triggering endothelin-1 signaling. Proc Natl Acad Sci U S A 2013; 110:E3225-34. [PMID: 23929777 DOI: 10.1073/pnas.1312933110] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
At our body surface, the epidermis absorbs UV radiation. UV overexposure leads to sunburn with tissue injury and pain. To understand how, we focus on TRPV4, a nonselective cation channel highly expressed in epithelial skin cells and known to function in sensory transduction, a property shared with other transient receptor potential channels. We show that following UVB exposure mice with induced Trpv4 deletions, specifically in keratinocytes, are less sensitive to noxious thermal and mechanical stimuli than control animals. Exploring the mechanism, we find that epidermal TRPV4 orchestrates UVB-evoked skin tissue damage and increased expression of the proalgesic/algogenic mediator endothelin-1. In culture, UVB causes a direct, TRPV4-dependent Ca(2+) response in keratinocytes. In mice, topical treatment with a TRPV4-selective inhibitor decreases UVB-evoked pain behavior, epidermal tissue damage, and endothelin-1 expression. In humans, sunburn enhances epidermal expression of TRPV4 and endothelin-1, underscoring the potential of keratinocyte-derived TRPV4 as a therapeutic target for UVB-induced sunburn, in particular pain.
Collapse
|