1
|
Frahm KS, Andersen OK, Arendt-Nielsen L, Gervasio S, Mørch CD. Topical capsaicin modulates the two-point discrimination threshold-Modulation depends on stimulation modality and intensity. Eur J Pain 2024; 28:1855-1865. [PMID: 39116004 DOI: 10.1002/ejp.4701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 06/07/2024] [Accepted: 07/04/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Spatial acuity concerns the ability to localize and discriminate sensory input and is often tested using the two-point discrimination threshold (2PDT). Sensitization of the pain system can affect the spatial acuity, but it is unclear how 2PDTs of different testing modalities are affected. The aim was to investigate if the 2PDTs for mechanical and heat stimulation at different intensities were modulated by topical capsaicin sensitization. METHODS 30 healthy subjects were divided into either a capsaicin or a placebo group. The 2PDT was tested using two different modalities, mechanical and thermal (laser) delivered at innocuous and noxious intensities. The 2PDT were determined at baseline and re-assessed 48 h later. In the follow-up session, the subjects either had a capsaicin patch (8%) or placebo patch placed in the testing area for 30 min before re-testing the 2PDT. RESULTS The 2PDT was highly dependent on stimulation modality and intensity. The lowest 2PDT was found for innocuous mechanical stimuli (40.0 mm, 95% CI 38.1-41.9 mm), and the highest 2PDT was found for innocuous thermal stimuli (81.7 mm, 95% CI 73.9-89.5 mm). Topical capsaicin generally increased the 2PDT, but this was only significant for innocuous mechanical stimuli. The perceived intensity of the stimuli was increased following capsaicin and was generally higher for noxious stimuli than for innocuous stimuli (ANOVA, p < 0.001). CONCLUSIONS This study showed that capsaicin provoked pain sensitization increased the 2PDT. The 2PDT tested using innocuous mechanical stimuli showed less variable results indicating that this test is most suitable to detect this aspect of spatial acuity. SIGNIFICANCE STATEMENT This study investigated how the two-point discrimination threshold (2PDT) can be modulated by topical capsaicin. The 2PDT was assessed for two different modalities (thermal and mechanical) and for two different intensities (innocuous and noxious) before and after capsaicin. The results showed that the 2PDT was generally impaired following capsaicin, but this was only significant for mechanical innocuous stimuli. Furthermore, it was shown that mechanical innocuous stimuli assessed the 2PDT with lower variability than other combinations.
Collapse
Affiliation(s)
- Ken Steffen Frahm
- Integrative Neuroscience Group, CNAP-Center for Neuroplasticity and Pain, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Ole Kæseler Andersen
- Integrative Neuroscience Group, CNAP-Center for Neuroplasticity and Pain, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Lars Arendt-Nielsen
- Translational Pain Biomarkers, CNAP-Center for Neuroplasticity and Pain, SMI®, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Department of Gastroenterology & Hepatology, Mech-Sense, Department of Clinical Medicine, Aalborg University Hospital, Aalborg, Denmark
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
| | - Sabata Gervasio
- Neural Engineering and Neurophysiology Group, SMI®, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Carsten Dahl Mørch
- Integrative Neuroscience Group, CNAP-Center for Neuroplasticity and Pain, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| |
Collapse
|
2
|
Vardigan JD, Pall PS, McDevitt DS, Huang C, Clements MK, Li Y, Kraus RL, Breslin MJ, Bungard CJ, Nemenov MI, Klukinov M, Burgey CS, Layton ME, Stachel SJ, Lange HS, Savitz AT, Santarelli VP, Henze DA, Uslaner JM. Analgesia and peripheral c-fiber modulation by selective Nav1.8 inhibition in rhesus. Pain 2024:00006396-990000000-00732. [PMID: 39382325 DOI: 10.1097/j.pain.0000000000003404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 08/01/2024] [Indexed: 10/10/2024]
Abstract
ABSTRACT Voltage-gated sodium (Nav) channels present untapped therapeutic value for better and safer pain medications. The Nav1.8 channel isoform is of particular interest because of its location on peripheral pain fibers and demonstrated role in rodent preclinical pain and neurophysiological assays. To-date, no inhibitors of this channel have been approved as drugs for treating painful conditions in human, possibly because of challenges in developing a sufficiently selective drug-like molecule with necessary potency not only in human but also across preclinical species critical to the preclinical development path of drug discovery. In addition, the relevance of rodent pain assays to the human condition is under increasing scrutiny as a number of mechanisms (or at the very least molecules) that are active in rodents have not translated to humans, and direct impact on pain fibers has not been confirmed in vivo. In this report, we have leveraged numerous physiological end points in nonhuman primates to evaluate the analgesic and pharmacodynamic activity of a novel, potent, and selective Nav1.8 inhibitor compound, MSD199. These pharmacodynamic biomarkers provide important confirmation of the in vivo impact of Nav1.8 inhibition on peripheral pain fibers in primates and have high translational potential to the clinical setting. These findings may thus greatly improve success of translational drug discovery efforts toward better and safer pain medications, as well as the understanding of primate biology of Nav1.8 inhibition broadly.
Collapse
Affiliation(s)
- Joshua D Vardigan
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Parul S Pall
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Dillon S McDevitt
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - ChienJung Huang
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Michelle K Clements
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Yuxing Li
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Richard L Kraus
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Michael J Breslin
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | | | | | | | - Chritopher S Burgey
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Mark E Layton
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Shawn J Stachel
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Henry S Lange
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Alan T Savitz
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Vincent P Santarelli
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Darrell A Henze
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| | - Jason M Uslaner
- Merck Sharp & Dohme LLC, a subsidiary of Merck & Co Inc, Rahway, NJ, United States
| |
Collapse
|
3
|
Hodkinson DJ, Drabek MM, Jung J, Lankappa ST, Auer DP. Theta Burst Stimulation of the Human Motor Cortex Modulates Secondary Hyperalgesia to Punctate Mechanical Stimuli. Neuromodulation 2024; 27:812-823. [PMID: 37952136 DOI: 10.1016/j.neurom.2023.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/19/2023] [Accepted: 10/03/2023] [Indexed: 11/14/2023]
Abstract
OBJECTIVES Many chronic pain conditions show evidence of dysregulated synaptic plasticity, including the development and maintenance of central sensitization. This provides a strong rationale for neuromodulation therapies for the relief of chronic pain. However, variability in responses and low fidelity across studies remain an issue for both clinical trials and pain management, demonstrating insufficient mechanistic understanding of effective treatment protocols. MATERIALS AND METHODS In a randomized counterbalanced crossover designed study, we evaluated two forms of patterned repetitive transcranial magnetic stimulation, known as continuous theta burst stimulation (TBS) and intermittent TBS, during normal and central sensitization states. Secondary hyperalgesia (a form of use-dependent central sensitization) was induced using a well-established injury-free pain model and assessed by standardized quantitative sensory testing involving light touch and pinprick pain thresholds in addition to stimulus-response functions. RESULTS We found that continuous TBS of the human motor cortex has a facilitatory (pronociceptive) effect on the magnitude of perceived pain to secondary hyperalgesia, which may rely on induction and expression of neural plasticity through heterosynaptic long-term potentiation-like mechanisms. CONCLUSIONS By defining the underlying mechanisms of TBS-driven synaptic plasticity in the nociceptive system, we offer new insight into disease mechanisms and provide targets for promoting functional recovery and repair in chronic pain. For clinical applications, this knowledge is critical for development of more efficacious and mechanisms-based neuromodulation protocols, which are urgently needed to address the chronic pain and opioid epidemics.
Collapse
Affiliation(s)
- Duncan J Hodkinson
- Division of Mental Health and Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK; Sir Peter Mansfield Imaging Center, School of Medicine, University of Nottingham, Nottingham, UK; National Institute for Health Research, Nottingham Biomedical Research Center, Queens Medical Center, Nottingham, UK.
| | - Marianne M Drabek
- Division of Mental Health and Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK; Sir Peter Mansfield Imaging Center, School of Medicine, University of Nottingham, Nottingham, UK; National Institute for Health Research, Nottingham Biomedical Research Center, Queens Medical Center, Nottingham, UK
| | - JeYoung Jung
- School of Psychology, University of Nottingham, Nottingham, UK
| | - Sudheer T Lankappa
- Nottinghamshire Healthcare National Health Service Foundation Trust, Nottingham, UK
| | - Dorothee P Auer
- Division of Mental Health and Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, UK; Sir Peter Mansfield Imaging Center, School of Medicine, University of Nottingham, Nottingham, UK; National Institute for Health Research, Nottingham Biomedical Research Center, Queens Medical Center, Nottingham, UK
| |
Collapse
|
4
|
Breslin MJ, Schubert JW, Wang D, Huang C, Clements MK, Li Y, Zhou X, Vardigan JD, Kraus RL, Santarelli VP, Uslaner JM, Coleman PJ, Stachel SJ. 2-Aminopyridines as Potent and Selective Na v1.8 Inhibitors Exhibiting Efficacy in a Nonhuman Primate Pain Model. ACS Med Chem Lett 2024; 15:917-923. [PMID: 38894930 PMCID: PMC11181479 DOI: 10.1021/acsmedchemlett.4c00103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Herein we describe the discovery of a 2-aminopyridine scaffold as a potent and isoform selective inhibitor of the Nav1.8 sodium channel. Parallel library synthesis, guided by in silico predictions, rapidly transformed initial hits into a novel 2-aminopyridine lead class possessing good ADME and pharmacokinetic profiles that were able to display activity in a clinically translatable nonhuman primate capsaicin-sensitized thermode pharmacodynamic assay. Progress toward the lead identification, optimization, and in vivo efficacy of these compounds will be discussed.
Collapse
Affiliation(s)
- Michael J. Breslin
- Discovery
Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Jeffrey W. Schubert
- Discovery
Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Deping Wang
- Modeling
and Informatics, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Chienjung Huang
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Michelle K. Clements
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Yuxing Li
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Xiaoping Zhou
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Joshua D. Vardigan
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Richard L. Kraus
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Vincent P. Santarelli
- Neuroscience
Biology Discovery, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Jason M. Uslaner
- Preclinical
and Translational Medicine Discovery, Merck
& Co., Inc., 770
Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Paul J. Coleman
- Discovery
Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| | - Shawn J. Stachel
- Discovery
Chemistry, Merck & Co., Inc., 770 Sumneytown Pike, P.O. Box 4, West Point, Pennsylvania 19486, United States
| |
Collapse
|
5
|
Galgiani JE, French MA, Morton SM. Acute pain impairs retention of locomotor learning. J Neurophysiol 2024; 131:678-688. [PMID: 38381551 PMCID: PMC11305642 DOI: 10.1152/jn.00343.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024] Open
Abstract
Despite abundant evidence that pain alters movement performance, considerably less is known about the potential effects of pain on motor learning. Some of the brain regions involved in pain processing are also responsible for specific aspects of motor learning, indicating that the two functions have the potential to interact, yet it is unclear if they do. In experiment 1, we compared the acquisition and retention of a novel locomotor pattern in young, healthy individuals randomized to either experience pain via capsaicin and heat applied to the lower leg during learning or no stimulus. On day 1, participants learned a new asymmetric walking pattern using distorted visual feedback, a paradigm known to involve mostly explicit re-aiming processes. Retention was tested 24 h later. Although there were no differences in day 1 acquisition between groups, individuals who experienced pain on day 1 demonstrated reduced retention on day 2. Furthermore, the degree of forgetting between days correlated with pain ratings during learning. In experiment 2, we examined the effects of a heat stimulus alone, which served as a control for (nonpainful) cutaneous stimulation, and found no effects on either acquisition or retention of learning. Thus, pain experienced during explicit, strategic locomotor learning interferes with motor memory consolidation processes and does so most likely through a pain mechanism and not an effect of distraction. These findings have important implications for understanding basic motor learning processes and for clinical rehabilitation, in which painful conditions are often treated through motor learning-based interventions.NEW & NOTEWORTHY Pain is a highly prevalent and burdensome experience that rehabilitation practitioners often treat using motor learning-based interventions. Here, we showed that experimental acute pain, but not a heat stimulus, during locomotor learning impaired 24-h retention of the newly learned walking pattern. The degree of retention loss was related to the perceived pain level during learning. These findings suggest important links between pain and motor learning that have significant implications for clinical rehabilitation.
Collapse
Affiliation(s)
- Jessica E Galgiani
- Department of Physical Therapy, University of Delaware, Newark, Delaware, United States
- Interdisciplinary Graduate Program in Biomechanics and Movement Science, University of Delaware, Newark, Delaware, United States
| | - Margaret A French
- Department of Physical Therapy, University of Delaware, Newark, Delaware, United States
- Interdisciplinary Graduate Program in Biomechanics and Movement Science, University of Delaware, Newark, Delaware, United States
| | - Susanne M Morton
- Department of Physical Therapy, University of Delaware, Newark, Delaware, United States
- Interdisciplinary Graduate Program in Biomechanics and Movement Science, University of Delaware, Newark, Delaware, United States
| |
Collapse
|
6
|
Wang W, Wang Y, Xiang L, Chen L, Yu L, Pan A, Tan J, Yuan Q. A Biomimetic Nociceptor Using Centrosymmetric Crystals for Machine Intelligence. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310555. [PMID: 38018790 DOI: 10.1002/adma.202310555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/25/2023] [Indexed: 11/30/2023]
Abstract
Pain sensation is a crucial aspect of perception in the body. Force-activated nociceptors encode electrochemical signals and yield multilevel information of pain, thus enabling smart feedback. Inspired by the natural template, multi-dimensional mechano-sensing materials provide promising approaches for biomimetic nociceptors in intelligent terminals. However, the reliance on non-centrosymmetric crystals has narrowed the range of these materials. Here centrosymmetric crystal Cr3+ -doped zinc gallogermanate (ZGGO:Cr) with multi-dimensional mechano-sensing is reported, eliminating the limitation of crystal structure. Under forces, ZGGO:Cr generates electrical signals imitating those of neuronal systems, and produces luminescence for spatial mapping of mechanical stimuli, suggesting a path toward bionic pain perception. On that basis, a wireless biomimetic nociceptor system is developed and a smart pain reflex in a robotic hand and robot-assisted biopsy surgery of rat and dog is achieved.
Collapse
Affiliation(s)
- Wenjie Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, Key Laboratory for Micro-Nano Physics and Technology of Hunan Province, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University, Changsha, 410082, China
| | - Yingfei Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, Key Laboratory for Micro-Nano Physics and Technology of Hunan Province, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University, Changsha, 410082, China
| | - Li Xiang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, Key Laboratory for Micro-Nano Physics and Technology of Hunan Province, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University, Changsha, 410082, China
| | - Long Chen
- Department of Computer and Information Science, Faculty of Science and Technology, University of Macau, Macau SAR, China
| | - Lilei Yu
- College of Chemistry and Molecular Sciences, Department of Cardiology, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| | - Anlian Pan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, Key Laboratory for Micro-Nano Physics and Technology of Hunan Province, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University, Changsha, 410082, China
| | - Jie Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, Key Laboratory for Micro-Nano Physics and Technology of Hunan Province, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University, Changsha, 410082, China
| | - Quan Yuan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, Key Laboratory for Micro-Nano Physics and Technology of Hunan Province, College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Hunan University, Changsha, 410082, China
- College of Chemistry and Molecular Sciences, Department of Cardiology, Institute of Molecular Medicine, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| |
Collapse
|
7
|
Wittkopf PG, Boye Larsen D, Gregoret L, Graven-Nielsen T. Disrupted Cortical Homeostatic Plasticity Due to Prolonged Capsaicin-induced Pain. Neuroscience 2023; 533:1-9. [PMID: 37774909 DOI: 10.1016/j.neuroscience.2023.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/08/2023] [Accepted: 09/20/2023] [Indexed: 10/01/2023]
Abstract
Homeostatic plasticity (HP) regulates cortical excitability (CE) stability but is disrupted in persistent pain conditions. This study investigated how prolonged experimental pain affects HP and if pain relief modulates disrupted HP. Twenty-four healthy participants were randomised into a PainRelief or NoPainRelief group and attended four sessions; two sessions on consecutive days, separated by two weeks. Transcranial magnetic stimulation motor-evoked potentials reflecting CE and quantitative sensory testing (QST) measures were recorded. A capsaicin (pain condition) or placebo (control condition) patch was applied to the hand. HP was induced by cathodal-cathodal transcranial direct current stimulation (HP1) with CE assessment before and after. The PainRelief group had ice applied to the patch, while the NoPainRelief group waited for five minutes; subsequently another HP induction (HP2) and CE assessment were performed. After 24 h with the patch on, HP induction (HP3), QST, and CE recordings were repeated. Capsaicin reduced CE and the pain condition showed disrupted homeostatic responses at all time points (HP1: showed CE inhibition instead of facilitation; HP2 & HP3: lack of CE facilitation). Conversely, homeostatic responses were induced at all time points for the placebo condition. Capsaicin pain disrupts HP which is not restored by ice-induced pain relief. Future research may explore the prevention of HP disruption by targeting capsaicin-induced nociception but not pain perception.
Collapse
Affiliation(s)
- Priscilla Geraldine Wittkopf
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Selma Lagerløfs Vej 249, 9260 Gistrup, Aalborg, Denmark
| | - Dennis Boye Larsen
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Selma Lagerløfs Vej 249, 9260 Gistrup, Aalborg, Denmark
| | - Luisina Gregoret
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Selma Lagerløfs Vej 249, 9260 Gistrup, Aalborg, Denmark
| | - Thomas Graven-Nielsen
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Selma Lagerløfs Vej 249, 9260 Gistrup, Aalborg, Denmark.
| |
Collapse
|
8
|
Campbell CM, Mun CJ, Hamilton KR, Bergeria CL, Huhn AS, Speed TJ, Vandrey R, Dunn KE. Within-subject, double-blind, randomized, placebo-controlled evaluation of combining the cannabinoid dronabinol and the opioid hydromorphone in adults with chronic pain. Neuropsychopharmacology 2023; 48:1630-1638. [PMID: 37202479 PMCID: PMC10516978 DOI: 10.1038/s41386-023-01597-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/20/2023]
Abstract
The potential synergistic effects of combining cannabinoids and opioids for analgesia has received considerable attention. No studies to date have evaluated this combination in patients with chronic pain. The present study aimed to evaluate the combined analgesic and drug effects of oral opioid (hydromorphone) and delta-9-tetrahydrocannabinol (dronabinol), as well as their effects on physical and cognitive functioning, and human abuse potential (HAP) outcomes among individuals with knee osteoarthritis (KOA). This was a within-subject, double-blind, randomized, placebo-controlled study. Participants (N = 37; 65% women; mean age = 62) diagnosed with knee osteoarthritis of ≥3/10 average pain intensity were included. Participants received (1) placebo-placebo, (2) hydromorphone (4 mg)-placebo; (3) dronabinol (10 mg)-placebo, and (4) hydromorphone (4 mg)-dronabinol (10 mg). Clinical and experimentally-induced pain, physical and cognitive function, subjective drug effects, HAP, adverse events, and pharmacokinetics were evaluated. No significant analgesic effects were observed for clinical pain severity or physical functioning across all drug conditions. Little enhancement of hydromorphone analgesia by dronabinol was observed on evoked pain indices. While subjective drug effects and some HAP ratings were increased in the combined drug condition, these were not significantly increased over the dronabinol alone condition. No serious adverse events were reported; hydromorphone produced more mild adverse events than placebo, but hydromorphone + dronabinol produced more moderate adverse events than both placebo and hydromorphone alone. Only hydromorphone impaired cognitive performance. Consistent with laboratory studies on healthy adults, the present study shows minimal benefit of combining dronabinol (10 mg) and hydromorphone (4 mg) for analgesia and improving physical functioning in adults with KOA.
Collapse
Affiliation(s)
- Claudia M Campbell
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Chung Jung Mun
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Arizona State University, Edson College of Nursing and Health Innovation, Phoenix, AZ, USA
| | - Katrina R Hamilton
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cecilia L Bergeria
- Behavioral Pharmacology Research Unit, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew S Huhn
- Behavioral Pharmacology Research Unit, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Traci J Speed
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ryan Vandrey
- Behavioral Pharmacology Research Unit, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kelly E Dunn
- Behavioral Pharmacology Research Unit, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Bested K, Jensen LM, Andresen T, Tarp G, Skovbjerg L, Johansen TS, Schmedes AV, Storgaard IK, Madsen JS, Werner MU, Bendiksen A. Low-dose naltrexone for treatment of pain in patients with fibromyalgia: a randomized, double-blind, placebo-controlled, crossover study. Pain Rep 2023; 8:e1080. [PMID: 38226027 PMCID: PMC10789452 DOI: 10.1097/pr9.0000000000001080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/26/2023] [Accepted: 04/15/2023] [Indexed: 01/17/2024] Open
Abstract
Introduction Fibromyalgia (FM) is a chronic fluctuating, nociplastic pain condition. Naltrexone is a µ-opioid-receptor antagonist; preliminary studies have indicated a pain-relieving effect of low-dose naltrexone (LDN) in patients with FM. The impetus for studying LDN is the assumption of analgesic efficacy and thus reduction of adverse effects seen from conventional pharmacotherapy. Objectives First, to examine if LDN is associated with analgesic efficacy compared with control in the treatment of patients with FM. Second, to ascertain the analgesic efficacy of LDN in an experimental pain model in patients with FM evaluating the competence of the descending inhibitory pathways compared with controls. Third, to examine the pharmacokinetics of LDN. Methods The study used a randomized, double-blind, placebo-controlled, crossover design and had a 3-phase setup. The first phase included baseline assessment and a treatment period (days -3 to 21), the second phase a washout period (days 22-32), and the third phase a baseline assessment followed by a treatment period (days 33-56). Treatment was with either LDN 4.5 mg or an inactive placebo given orally once daily. The primary outcomes were Fibromyalgia Impact Questionnaire revised (FIQR) scores and summed pain intensity ratings (SPIR). Results Fifty-eight patients with FM were randomized. The median difference (IQR) for FIQR scores between LDN and placebo treatment was -1.65 (18.55; effect size = 0.15; P = 0.3). The median difference for SPIR scores was -0.33 (6.33; effect size = 0.13; P = 0.4). Conclusion Outcome data did not indicate any clinically relevant analgesic efficacy of the LDN treatment in patients with FM.
Collapse
Affiliation(s)
- Kirsten Bested
- Multidisciplinary Pain Clinic, Friklinikken, Grindsted, Denmark
| | - Lotte M. Jensen
- Multidisciplinary Pain Clinic, Friklinikken, Grindsted, Denmark
| | - Trine Andresen
- Molecular Diagnostics and Clinical Research Unit, Hospital Sonderjylland, Aabendraa, Denmark
| | - Grete Tarp
- Multidisciplinary Pain Clinic, Friklinikken, Grindsted, Denmark
| | - Louise Skovbjerg
- Multidisciplinary Pain Center, Neuroscience Center, Rigshospitalet, Copenhagen University Hospitals, Copenhagen, Denmark
| | | | - Anne V. Schmedes
- Department of Clinical Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Ida K. Storgaard
- Department of Drug Design and Pharmacology, Copenhagen University Hospitals, Copenhagen, Denmark
| | - Jonna S. Madsen
- Department of Clinical Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Mads U. Werner
- Multidisciplinary Pain Center, Neuroscience Center, Rigshospitalet, Copenhagen University Hospitals, Copenhagen, Denmark
| | | |
Collapse
|
10
|
Lötsch J, Mayer B, Kringel D. Machine learning analysis predicts a person's sex based on mechanical but not thermal pain thresholds. Sci Rep 2023; 13:7332. [PMID: 37147321 PMCID: PMC10163041 DOI: 10.1038/s41598-023-33337-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/11/2023] [Indexed: 05/07/2023] Open
Abstract
Sex differences in pain perception have been extensively studied, but precision medicine applications such as sex-specific pain pharmacology have barely progressed beyond proof-of-concept. A data set of pain thresholds to mechanical (blunt and punctate pressure) and thermal (heat and cold) stimuli applied to non-sensitized and sensitized (capsaicin, menthol) forearm skin of 69 male and 56 female healthy volunteers was analyzed for data structures contingent with the prior sex structure using unsupervised and supervised approaches. A working hypothesis that the relevance of sex differences could be approached via reversibility of the association, i.e., sex should be identifiable from pain thresholds, was verified with trained machine learning algorithms that could infer a person's sex in a 20% validation sample not seen to the algorithms during training, with balanced accuracy of up to 79%. This was only possible with thresholds for mechanical stimuli, but not for thermal stimuli or sensitization responses, which were not sufficient to train an algorithm that could assign sex better than by guessing or when trained with nonsense (permuted) information. This enabled the translation to the molecular level of nociceptive targets that convert mechanical but not thermal information into signals interpreted as pain, which could eventually be used for pharmacological precision medicine approaches to pain. By exploiting a key feature of machine learning, which allows for the recognition of data structures and the reduction of information to the minimum relevant, experimental human pain data could be characterized in a way that incorporates "non" logic that could be translated directly to the molecular pharmacological level, pointing toward sex-specific precision medicine for pain.
Collapse
Affiliation(s)
- Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany.
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596, Frankfurt, Germany.
| | - Benjamin Mayer
- Institute of Clinical Pharmacology, Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Dario Kringel
- Institute of Clinical Pharmacology, Goethe-University, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| |
Collapse
|
11
|
Jang W, Oh M, Cho EH, Baek M, Kim C. Drosophila pain sensitization and modulation unveiled by a novel pain model and analgesic drugs. PLoS One 2023; 18:e0281874. [PMID: 36795675 PMCID: PMC9934396 DOI: 10.1371/journal.pone.0281874] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 02/01/2023] [Indexed: 02/17/2023] Open
Abstract
In mammals, pain is regulated by the combination of an ascending stimulating and descending inhibitory pain pathway. It remains an intriguing question whether such pain pathways are of ancient origin and conserved in invertebrates. Here we report a new Drosophila pain model and use it to elucidate the pain pathways present in flies. The model employs transgenic flies expressing the human capsaicin receptor TRPV1 in sensory nociceptor neurons, which innervate the whole fly body, including the mouth. Upon capsaicin sipping, the flies abruptly displayed pain-related behaviors such as running away, scurrying around, rubbing vigorously, and pulling at their mouth parts, suggesting that capsaicin stimulated nociceptors in the mouth via activating TRPV1. When reared on capsaicin-containing food, the animals died of starvation, demonstrating the degree of pain experienced. This death rate was reduced by treatment both with NSAIDs and gabapentin, analgesics that inhibit the sensitized ascending pain pathway, and with antidepressants, GABAergic agonists, and morphine, analgesics that strengthen the descending inhibitory pathway. Our results suggest Drosophila to possess intricate pain sensitization and modulation mechanisms similar to mammals, and we propose that this simple, non-invasive feeding assay has utility for high-throughput evaluation and screening of analgesic compounds.
Collapse
Affiliation(s)
- Wijeong Jang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Myungsok Oh
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Eun-Hee Cho
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Minwoo Baek
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Changsoo Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
- * E-mail:
| |
Collapse
|
12
|
Alhajri N, Boudreau SA, Graven-Nielsen T. Decreased Default Mode Network Connectivity Following 24 Hours of Capsaicin-induced Pain Persists During Immediate Pain Relief and Facilitation. THE JOURNAL OF PAIN 2022; 24:796-811. [PMID: 36521671 DOI: 10.1016/j.jpain.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022]
Abstract
Prolonged experimental pain models can help assess cortical mechanisms underlying the transition from acute to chronic pain such as resting-state functional connectivity (rsFC), especially in early stages. This crossover study determined the effects of 24-hour-capsaicin-induced pain on the default mode network rsFC, a major network in the dynamic pain connectome. Electroencephalographic rsFC measured by Granger causality was acquired from 24 healthy volunteers (12 women) at baseline, 1hour, and 24hours following the application of a control or capsaicin patch on the right forearm. The control patch was received maximum 1 week before the capsaicin patch. Following 24hours, the patch was cooled and later heated to assess rsFC changes in response to pain relief and facilitation, respectively. Compared to baseline, decreased rsFC at alpha oscillations (8-10Hz) was found following 1hour and 24hours of capsaicin application for connections projecting from medial prefrontal cortex (mPFC) and right angular gyrus (rAG) but not left angular gyrus (lAG) or posterior cingulate cortex (PCC): mPFC-PCC (1hour:P < .001, 24hours:P = .002), mPFC-rAG (1hour:P < .001, 24hours:P = .001), rAG-mPFC (1hour:P < .001, 24hours:P = .001), rAG-PCC (1hour:P < .001, 24hours:P = .004). Comparable decreased rsFC following 1hour and 24hours (P≤0.008) was found at beta oscillations, however, decreased projections from PCC were also found: PCC-rAG (P≤0.005) and PCC-lAG (P≤0.006). Pain NRS scores following 24hours (3.7±0.4) was reduced by cooling (0.3±0.1, P = .004) and increased by heating (4.8±0.6, P = .016). However, neither cooling nor heating altered rsFC. This study shows that 24hours of experimental pain induces a robust decrease in DMN connectivity that persists during pain relief or facilitation suggesting a possible shift to attentional and emotional processing in persistent pain. PERSPECTIVE: This article shows decreased DMN connectivity that might reflect possible attentional and emotional changes during acute and prolonged pain. Understanding these changes could potentially help clinicians in developing therapeutic methods that can better target these attentional and emotional processes before developing into more persistent states.
Collapse
Affiliation(s)
- Najah Alhajri
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Shellie Ann Boudreau
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Thomas Graven-Nielsen
- Center for Neuroplasticity and Pain (CNAP), Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
13
|
The Sleep-Reward-Pain Pathway Model: an Integrative Review. CURRENT SLEEP MEDICINE REPORTS 2022. [DOI: 10.1007/s40675-022-00232-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Sympathetic skin response as an objective tool to estimate stimulus-associated arousal in a human model of hyperalgesia. Neurophysiol Clin 2022; 52:436-445. [DOI: 10.1016/j.neucli.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 08/02/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
|
15
|
Tonic pain alters functional connectivity of the descending pain modulatory network involving amygdala, periaqueductal gray, parabrachial nucleus and anterior cingulate cortex. Neuroimage 2022; 256:119278. [PMID: 35523367 PMCID: PMC9250649 DOI: 10.1016/j.neuroimage.2022.119278] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 04/07/2022] [Accepted: 05/02/2022] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION Resting state functional connectivity (FC) is widely used to assess functional brain alterations in patients with chronic pain. However, reports of FC accompanying tonic pain in pain-free persons are rare. A network we term the Descending Pain Modulatory Network (DPMN) is implicated in healthy and pathologic pain modulation. Here, we evaluate the effect of tonic pain on FC of specific nodes of this network: anterior cingulate cortex (ACC), amygdala (AMYG), periaqueductal gray (PAG), and parabrachial nuclei (PBN). METHODS In 50 pain-free participants (30F), we induced tonic pain using a capsaicin-heat pain model. functional MRI measured resting BOLD signal during pain-free rest with a 32°C thermode and then tonic pain where participants experienced a previously warm temperature combined with capsaicin. We evaluated FC from ACC, AMYG, PAG, and PBN with correlation of self-report pain intensity during both states. We hypothesized tonic pain would diminish FC dyads within the DPMN. RESULTS Of all hypothesized FC dyads, only PAG and subgenual ACC was weakly altered during pain (F=3.34; p=0.074; pain-free>pain d=0.25). After pain induction sACC-PAG FC became positively correlated with pain intensity (R=0.38; t=2.81; p=0.007). Right PBN-PAG FC during pain-free rest positively correlated with subsequently experienced pain (R=0.44; t=3.43; p=0.001). During pain, this connection's FC was diminished (paired t=-3.17; p=0.0026). In whole-brain analyses, during pain-free rest, FC between left AMYG and right superior parietal lobule and caudate nucleus were positively correlated with subsequent pain. During pain, FC between left AMYG and right inferior temporal gyrus negatively correlated with pain. Subsequent pain positively correlated with right AMYG FC with right claustrum; right primary visual cortex and right temporo-occipitoparietal junction Conclusion: We demonstrate sACC-PAG tonic pain FC positively correlates with experienced pain and resting right PBN-PAG FC correlates with subsequent pain and is diminished during tonic pain. Finally, we reveal PAG- and right AMYG-anchored networks which correlate with subsequently experienced pain intensity. Our findings suggest specific connectivity patterns within the DPMN at rest are associated with subsequently experienced pain and modulated by tonic pain. These nodes and their functional modulation may reveal new therapeutic targets for neuromodulation or biomarkers to guide interventions.
Collapse
|
16
|
van der Schaaf ME, Schmidt K, Kaur J, Gamer M, Wiech K, Forkmann K, Bingel U. Acquisition learning is stronger for aversive than appetitive events. Commun Biol 2022; 5:302. [PMID: 35379893 PMCID: PMC8979974 DOI: 10.1038/s42003-022-03234-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 03/04/2022] [Indexed: 11/25/2022] Open
Abstract
Appetitive and aversive learning are both key building blocks of adaptive behavior, yet knowledge regarding their differences is sparse. Using a capsaicin heat pain model in 36 healthy participants, this study directly compared the acquisition and extinction of conditioned stimuli (CS) predicting pain exacerbation and relief. Valence ratings show stronger acquisition during aversive compared to appetitive learning, but no differences in extinction. Skin conductance responses and contingency ratings confirmed these results. Findings were unrelated to individual differences in pain sensitivity or psychological factors. Our results support the notion of an evolutionarily hardwired preponderance to acquire aversive rather than appetitive cues as is protective for acute aversive states such as pain but may contribute to the development and maintenance of clinical conditions such as chronic pain, depression or anxiety disorders. In this study, acquisition and extinction of conditioned stimuli are compared in a capsaicin heat pain paradigm in healthy human participants. The results demonstrate a preponderance to acquire aversive rather than appetitive cues, which could initially have a protective function but eventually contribute to the maintenance of chronic pain.
Collapse
Affiliation(s)
- Marieke E van der Schaaf
- Radboud University Medical Centre, Department of Psychiatry, 6525 GA, Nijmegen, The Netherlands.,Radboud University, Donders Institute for Brain Behaviour and Cognition, Centre for Cognitive Neuroimaging, Nijmegen, The Netherlands
| | - Katharina Schmidt
- Department of Neurology, Center for Translational Neuro- and Behavioural Sciences, University Medicine Essen, Essen, Germany.
| | - Jaspreet Kaur
- Department of Neurology, Center for Translational Neuro- and Behavioural Sciences, University Medicine Essen, Essen, Germany
| | - Matthias Gamer
- Department of Psychology, University of Würzburg, Würzburg, Germany
| | - Katja Wiech
- Wellcome Centre for Integrative Neuroimaging (WIN), Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Katarina Forkmann
- Department of Neurology, Center for Translational Neuro- and Behavioural Sciences, University Medicine Essen, Essen, Germany
| | - Ulrike Bingel
- Department of Neurology, Center for Translational Neuro- and Behavioural Sciences, University Medicine Essen, Essen, Germany
| |
Collapse
|
17
|
Bertrand-Charette M, Jeffrey-Gauthier R, Roy JS, Bouyer LJ. Gait Adaptation to a Phase-Specific Nociceptive Electrical Stimulation Applied at the Ankle: A Model to Study Musculoskeletal-Like Pain. Front Hum Neurosci 2022; 15:762450. [PMID: 34975433 PMCID: PMC8718644 DOI: 10.3389/fnhum.2021.762450] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 11/30/2021] [Indexed: 11/15/2022] Open
Abstract
Introduction: Lower limb pain, whether induced experimentally or as a result of a musculoskeletal injury, can impair motor control, leading to gait adaptations such as increased muscle stiffness or modified load distribution around joints. These adaptations may initially reduce pain but can also lead to longer-term maladaptive plasticity and to the development of chronic pain. In humans, many current experimental musculoskeletal-like pain models are invasive, and most don’t accurately reproduce the movement-related characteristics of musculoskeletal pain. The main objective of this study was to measure pain adaptation strategies during gait of a musculoskeletal-like experimental pain protocol induced by phase-specific, non-invasive electrical stimulation. Methods: Sixteen healthy participants walked on a treadmill at 4 km/h for three consecutive periods (BASELINE, PAIN, and POST-PAIN). Painful electrical stimulations were delivered at heel strike for the duration of heel contact (HC) using electrodes placed around the right lateral malleolus to mimic ankle sprains. Gait adaptations were quantified bilaterally using instrumented pressure-sensitive insoles. One-way ANOVAs and group time course analyses were performed to characterize the impact of electrical stimulation on heel and forefoot contact pressure and contact duration. Results: During the first few painful strides, peak HC pressure decreased on the painful side (8.6 ± 1.0%, p < 0.0001) and increased on the non-stimulated side (11.9 ± 0.9%, p < 0.0001) while HC duration was significantly reduced bilaterally (painful: 12.1 ± 0.9%, p < 0.0001; non-stimulated: 4.8 ± 0.8%, p < 0.0001). No clinically meaningful modifications were observed for the forefoot. One minute after the onset of painful stimulation, perceived pain levels stabilized and peak HC pressure remained significantly decreased on the painful side, while the other gait adaptations returned to pre-stimulation values. Discussion: These results demonstrate that a non-invasive, phase-specific pain can produce a stable painful gait pattern. Therefore, this protocol will be useful to study musculoskeletal pain locomotor adaptation strategies under controlled conditions.
Collapse
Affiliation(s)
- Michaël Bertrand-Charette
- Center for Interdisciplinary Research in Rehabilitation and Social Integration, Quebec City, QC, Canada
| | - Renaud Jeffrey-Gauthier
- Center for Interdisciplinary Research in Rehabilitation and Social Integration, Quebec City, QC, Canada
| | - Jean-Sébastien Roy
- Center for Interdisciplinary Research in Rehabilitation and Social Integration, Quebec City, QC, Canada.,Department of Rehabilitation, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| | - Laurent J Bouyer
- Center for Interdisciplinary Research in Rehabilitation and Social Integration, Quebec City, QC, Canada.,Department of Rehabilitation, Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
18
|
Springborg AD, Wessel CR, Andersen LPK, Werner MU. Methodology and applicability of the human contact burn injury model: A systematic review. PLoS One 2021; 16:e0254790. [PMID: 34329326 PMCID: PMC8323928 DOI: 10.1371/journal.pone.0254790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 07/06/2021] [Indexed: 11/24/2022] Open
Abstract
The contact burn injury model is an experimental contact thermode-based physiological pain model primarily applied in research of drug efficacy in humans. The employment of the contact burn injury model across studies has been inconsistent regarding essential methodological variables, challenging the validity of the model. This systematic review analyzes methodologies, outcomes, and research applications of the contact burn injury model. Based on these results, we propose an improved contact burn injury testing paradigm. A literature search was conducted (15-JUL-2020) using PubMed, EMBASE, Web of Science, and Google Scholar. Sixty-four studies were included. The contact burn injury model induced consistent levels of primary and secondary hyperalgesia. However, the analyses revealed variations in the methodology of the contact burn injury heating paradigm and the post-burn application of test stimuli. The contact burn injury model had limited testing sensitivity in demonstrating analgesic efficacy. There was a weak correlation between experimental and clinical pain intensity variables. The data analysis was limited by the methodological heterogenicity of the different studies and a high risk of bias across the studies. In conclusion, although the contact burn injury model provides robust hyperalgesia, it has limited efficacy in testing analgesic drug response. Recommendations for future use of the model are being provided, but further research is needed to improve the sensitivity of the contact burn injury method. The protocol for this review has been published in PROSPERO (ID: CRD42019133734).
Collapse
Affiliation(s)
- Anders Deichmann Springborg
- Department of Anesthesia, Multidisciplinary Pain Center, Pain and Respiratory Support, Neuroscience Center, Copenhagen University Hospital, Copenhagen, Denmark
- * E-mail:
| | - Caitlin Rae Wessel
- Department of Physiology, University of Kentucky, Lexington, KY, United States of America
| | - Lars Peter Kloster Andersen
- Department of Anaesthesia and Intensive Care, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Mads Utke Werner
- Department of Anesthesia, Multidisciplinary Pain Center, Pain and Respiratory Support, Neuroscience Center, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
19
|
Dunn KE, Bergeria CL, Huhn AS, Speed TJ, Mun CJ, Vandrey R, Campbell CM. Within-subject, double-blinded, randomized, and placebo-controlled evaluation of the combined effects of the cannabinoid dronabinol and the opioid hydromorphone in a human laboratory pain model. Neuropsychopharmacology 2021; 46:1451-1459. [PMID: 33879842 PMCID: PMC8055479 DOI: 10.1038/s41386-021-01007-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/04/2021] [Accepted: 03/24/2021] [Indexed: 02/02/2023]
Abstract
This Phase II study evaluated analgesia, abuse liability, and cognitive performance of hydromorphone and oral delta-9-tetrahydrocannabinol (THC; dronabinol) using a within-subject, double-blind, randomized, placebo-controlled, human laboratory trial. Healthy adults (N = 29) with no history of drug use disorder received combinations of placebo, hydromorphone (4 mg; oral), and dronabinol (2.5 mg, 5.0 mg, 10 mg; oral). Primary outcomes were quantitative sensory testing (QST) measures of acute (thermal, pressure pain; thermal, punctate probe temporal summation; cold pressor; conditioned pain modulation) and chronic pain (capsaicin 10% topical cream with thermal rekindling), measures of drug abuse liability, cognitive functioning, and adverse events. Subgroup analyses were conducted within opioid-responders (endorsed >20 on a Drug Effect visual analog scale during the hydromorphone-only condition) and nonresponders. A consistent dose-effect relationship of dronabinol on hydromorphone across all measures was not observed. Analgesia only improved in the hydromorphone + dronabinol 2.5 mg condition. Hydromorphone + dronabinol 2.5 mg showed the lowest and hydromorphone+dronabinol 5 mg showed the highest risk for abuse. Hydromorphone+dronabinol 10 mg produced a high rate of dysphoric effects, and hydromorphone+dronabinol 5 mg and hydromorphone + dronabinol 10 mg produced AEs. Subgroup analyses showed subjective effects and abuse risk was increased among opioid responders and largely absent among nonresponders. Overall, only hydromorphone+dronabinol 2.5 mg modestly enhanced hydromorphone-based analgesia and hydromorphone + dronabinol 5 mg and 10 mg increased risk for abuse and AEs. These data can help inform opioid-sparing efforts in clinical pain populations. Demonstration that potential opioid effects varied as a function of participant opioid sensitivity (e.g., responder status) is a novel finding that warrants additional research.
Collapse
Affiliation(s)
- Kelly E. Dunn
- grid.21107.350000 0001 2171 9311Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Cecilia L. Bergeria
- grid.21107.350000 0001 2171 9311Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Andrew S. Huhn
- grid.21107.350000 0001 2171 9311Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Traci J. Speed
- grid.21107.350000 0001 2171 9311Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Chung Jung Mun
- grid.21107.350000 0001 2171 9311Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Ryan Vandrey
- grid.21107.350000 0001 2171 9311Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Claudia M. Campbell
- grid.21107.350000 0001 2171 9311Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
20
|
Modality-specific facilitation of noninjurious sharp mechanical pain by topical capsaicin. Pain 2021; 162:275-286. [PMID: 32701656 DOI: 10.1097/j.pain.0000000000002020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We had previously shown that a "blunt blade" stimulator can mimic the noninjurious strain phase of incisional pain, but not its sustained duration. Here, we tested whether acute sensitization of the skin with topical capsaicin can add the sustained phase to this noninvasive surrogate model of intraoperative pain. Altogether, 110 healthy volunteers (55 male and 55 female; 26 ± 5 years) participated in several experiments using the "blunt blade" (0.25 × 4 mm) on normal skin (n = 36) and on skin pretreated by a high-concentration capsaicin patch (8%, Qutenza; n = 36). These data were compared with an experimental incision (n = 40) using quantitative and qualitative pain ratings by numerical rating scale and SES Pain Perception Scale descriptors. Capsaicin sensitization increased blade-induced pain magnitude and duration significantly (both P < 0.05), but it failed to fully match the sustained duration of incisional pain. In normal skin, the SES pattern of pain qualities elicited by the blade matched incision in pain magnitude and pattern of pain descriptors. In capsaicin-treated skin, the blade acquired a significant facilitation only of the perceived heat pain component (P < 0.001), but not of mechanical pain components. Thus, capsaicin morphed the descriptor pattern of the blade to become more capsaicin-like, which is probably explained best by peripheral sensitization of the TRPV1 receptor. Quantitative sensory testing in capsaicin-sensitized skin revealed hyperalgesia to heat and pressure stimuli, and loss of cold and cold pain sensitivity. These findings support our hypothesis that the blade models the early tissue-strain-related mechanical pain phase of surgical incisions.
Collapse
|
21
|
Quesada C, Kostenko A, Ho I, Leone C, Nochi Z, Stouffs A, Wittayer M, Caspani O, Brix Finnerup N, Mouraux A, Pickering G, Tracey I, Truini A, Treede RD, Garcia-Larrea L. Human surrogate models of central sensitization: A critical review and practical guide. Eur J Pain 2021; 25:1389-1428. [PMID: 33759294 PMCID: PMC8360051 DOI: 10.1002/ejp.1768] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/17/2021] [Accepted: 03/21/2021] [Indexed: 12/11/2022]
Abstract
Background As in other fields of medicine, development of new medications for management of neuropathic pain has been difficult since preclinical rodent models do not necessarily translate to the clinics. Aside from ongoing pain with burning or shock‐like qualities, neuropathic pain is often characterized by pain hypersensitivity (hyperalgesia and allodynia), most often towards mechanical stimuli, reflecting sensitization of neural transmission. Data treatment We therefore performed a systematic literature review (PubMed‐Medline, Cochrane, WoS, ClinicalTrials) and semi‐quantitative meta‐analysis of human pain models that aim to induce central sensitization, and generate hyperalgesia surrounding a real or simulated injury. Results From an initial set of 1569 reports, we identified and analysed 269 studies using more than a dozen human models of sensitization. Five of these models (intradermal or topical capsaicin, low‐ or high‐frequency electrical stimulation, thermode‐induced heat‐injury) were found to reliably induce secondary hyperalgesia to pinprick and have been implemented in multiple laboratories. The ability of these models to induce dynamic mechanical allodynia was however substantially lower. The proportion of subjects who developed hypersensitivity was rarely provided, giving rise to significant reporting bias. In four of these models pharmacological profiles allowed to verify similarity to some clinical conditions, and therefore may inform basic research for new drug development. Conclusions While there is no single “optimal” model of central sensitization, the range of validated and easy‐to‐use procedures in humans should be able to inform preclinical researchers on helpful potential biomarkers, thereby narrowing the translation gap between basic and clinical data. Significance Being able to mimic aspects of pathological pain directly in humans has a huge potential to understand pathophysiology and provide animal research with translatable biomarkers for drug development. One group of human surrogate models has proven to have excellent predictive validity: they respond to clinically active medications and do not respond to clinically inactive medications, including some that worked in animals but failed in the clinics. They should therefore inform basic research for new drug development.
Collapse
Affiliation(s)
- Charles Quesada
- NeuroPain lab, Lyon Centre for Neuroscience Inserm U1028, Lyon, France.,Pain Center Neurological Hospital (CETD), Hospices Civils de Lyon, Lyon, France
| | - Anna Kostenko
- Department of Neurophysiology, Mannheim center for Translational Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Idy Ho
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Caterina Leone
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Zahra Nochi
- Danish Pain Research Center, Dept of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Alexandre Stouffs
- Institute of Neuroscience (IoNS), Université Catholique de Louvain (UCLouvain), Ottignies-Louvain-la-Neuve, Belgium
| | - Matthias Wittayer
- Department of Neurophysiology, Mannheim center for Translational Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Ombretta Caspani
- Department of Neurophysiology, Mannheim center for Translational Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Nanna Brix Finnerup
- Danish Pain Research Center, Dept of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - André Mouraux
- Institute of Neuroscience (IoNS), Université Catholique de Louvain (UCLouvain), Ottignies-Louvain-la-Neuve, Belgium
| | | | - Irene Tracey
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Andrea Truini
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Rolf-Detlef Treede
- Department of Neurophysiology, Mannheim center for Translational Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Luis Garcia-Larrea
- NeuroPain lab, Lyon Centre for Neuroscience Inserm U1028, Lyon, France.,Pain Center Neurological Hospital (CETD), Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
22
|
The metabotropic glutamate receptor 5 negative allosteric modulator fenobam: pharmacokinetics, side effects, and analgesic effects in healthy human subjects. Pain 2021; 161:135-146. [PMID: 31568235 DOI: 10.1097/j.pain.0000000000001695] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Metabotropic glutamate receptor 5 (mGlu5) has been shown to modulate nociception in animals, but no mGlu5 antagonists have been developed commercially as analgesics. The mGlu5 antagonist fenobam [N-(3-chlorophenyl)-N'-(4,5-dihydro-1-methyl-4-oxo-1H-imidazole-2-yl)urea] was originally evaluated for development as a nonbenzodiazepine anxiolytic. Fenobam is analgesic in numerous mouse pain models, acting exclusively through mGlu5 blockade. Furthermore, fenobam showed no signs of analgesic tolerance with up to 2 weeks of daily dosing in mice. Analgesic effects of fenobam in humans have not been reported. The purpose of this investigation was to evaluate fenobam pharmacokinetics and analgesic effects in humans. We first evaluated single-dose oral fenobam disposition in a parallel-group dose-escalation study in healthy volunteers. A second investigation tested the analgesic effects of fenobam in an established experimental human pain model of cutaneous sensitization using capsaicin cream and heat, in a double-blind placebo-controlled study. The primary outcome measure was the area of hyperalgesia and allodynia around the area applied with heat/capsaicin. Secondary outcome measures included nociception, measured as pain rating on a visual analog scale, heat pain detection threshold, and effects on cognition and mood. Fenobam plasma exposures showed considerable interindividual variability and were not linear with dose. Fenobam reduced sensitization vs placebo at a single timepoint (peak plasma concentration); we found no other difference between fenobam and placebo. Our results suggest highly variable fenobam disposition and minimal analgesic effects at the dose tested. We suggest that future studies testing analgesic effects of mGlu5 blockade are warranted, but such studies should use molecules with improved pharmacokinetic profiles.
Collapse
|
23
|
Scheuren PS, Rosner J, Curt A, Hubli M. Pain-autonomic interaction: A surrogate marker of central sensitization. Eur J Pain 2020; 24:2015-2026. [PMID: 32794307 DOI: 10.1002/ejp.1645] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 11/07/2022]
Abstract
BACKGROUND Central sensitization represents a key pathophysiological mechanism underlying the development of neuropathic pain, often manifested clinically as mechanical allodynia and hyperalgesia. Adopting a mechanism-based treatment approach relies highly on the ability to assess the presence of central sensitization. The aim of the study was to investigate potential pain-autonomic readouts to operationalize experimentally induced central sensitization in the area of secondary hyperalgesia. METHODS Pinprick evoked potentials (PEPs) and sympathetic skin responses (SSRs) were recorded in 20 healthy individuals. Three blocks of PEP and SSR recordings were performed before and after heat-induced secondary hyperalgesia. All measurements were also performed before and after a control condition. Multivariate analyses were performed using linear mixed-effect regression models to examine the effect of experimentally induced central sensitization on PEP and SSR parameters (i.e. amplitudes, latencies and habituation) and on pinprick pain ratings. RESULTS The noxious heat stimulation induced robust mechanical hyperalgesia with a significant increase in PEP and SSR amplitudes (p < 0.001) in the area of secondary hyperalgesia. Furthermore, PEP and SSR habituation were reduced (p < 0.001) after experimentally induced central sensitization. CONCLUSIONS The findings demonstrate that combined recordings of PEPs and SSRs are sensitive to objectify experimentally induced central sensitization and may have a great potential to reveal its presence in clinical pain conditions. Corroborating current pain phenotyping with pain-autonomic markers has the potential to unravel central sensitization along the nociceptive neuraxis and might provide a framework for mechanistically founded therapies. SIGNIFICANCE Our findings provide evidence that combined recordings of sympathetic skin responses (SSRs) and pinprick evoked potentials (PEPs) might be able to unmask central sensitization induced through a well-established experimental pain model in healthy individuals. As such, these novel readouts of central sensitization might attain new insights towards complementing clinical pain phenotyping.
Collapse
Affiliation(s)
- Paulina S Scheuren
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Jan Rosner
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland.,Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Armin Curt
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Michèle Hubli
- Spinal Cord Injury Center, Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
24
|
Smith MT, Remeniuk B, Finan PH, Speed TJ, Tompkins DA, Robinson M, Gonzalez K, Bjurstrom MF, Irwin MR. Sex differences in measures of central sensitization and pain sensitivity to experimental sleep disruption: implications for sex differences in chronic pain. Sleep 2020; 42:5146314. [PMID: 30371854 DOI: 10.1093/sleep/zsy209] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Indexed: 01/11/2023] Open
Abstract
Study Objectives Females demonstrate heightened central sensitization (CS), a risk factor for chronic pain characterized by enhanced responsivity of central nervous system nociceptors to normal or subthreshold input. Sleep disruption increases pain sensitivity, but sex has rarely been evaluated as a moderator and few experiments have measured CS. We evaluated whether two nights of sleep disruption alter CS measures of secondary hyperalgesia and mechanical temporal summation in a sex-dependent manner. We also evaluated differences in measures of pain sensitivity. Methods Seventy-nine healthy adults (female n = 46) participated in a randomized crossover experiment comparing two consecutive nights of eight pseudorandomly distributed forced awakenings (FA [-200 min sleep time]) against two nights of undisturbed sleep (US). We conducted sensory testing the mornings following Night 2; the heat-capsaicin pain model was used to induce secondary hyperalgesia. Results FA reduced total sleep time (REM and NREM Stage 3) more profoundly in males. We observed divergent, sex-dependent effects of FA on secondary hyperalgesia and temporal summation. FA significantly increased secondary hyperalgesia in males and significantly increased temporal summation in females. Sex differences were not attributable to differential sleep loss in males. FA also significantly reduced heat-pain threshold and cold pressor pain tolerance, independently of sex. Conclusions Sleep disruption enhances different pain facilitatory measures of CS in males and females suggesting that sleep disturbance may increase risk for chronic pain in males and females via distinct pathways. Findings have implications for understanding sex differences in chronic pain and investigating sleep in chronic pain prevention efforts.
Collapse
Affiliation(s)
- Michael T Smith
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD
| | - Bethany Remeniuk
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD
| | - Patrick H Finan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD
| | - Traci J Speed
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD
| | - D Andrew Tompkins
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD.,Department of Psychiatry, UCSF School of Medicine, San Francisco, CA
| | - Mercedes Robinson
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD
| | - Kaylin Gonzalez
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, School of Medicine, Baltimore, MD
| | - Martin F Bjurstrom
- Department of Psychiatry and Behavioral Sciences, Cousins Center for Psychoneuroimmunology, UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA
| | - Michael R Irwin
- Department of Psychiatry and Behavioral Sciences, Cousins Center for Psychoneuroimmunology, UCLA Semel Institute for Neuroscience and Human Behavior, Los Angeles, CA
| |
Collapse
|
25
|
Hoeger Bement M, Petersen KK, Sørensen LB, Andersen HH, Graven‐Nielsen T. Temporal aspects of endogenous pain modulation during a noxious stimulus prolonged for 1 day. Eur J Pain 2020; 24:752-760. [DOI: 10.1002/ejp.1523] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 01/29/2023]
Affiliation(s)
- Marie Hoeger Bement
- Center for Neuroplasticity and Pain (CNAP) Aalborg University Aalborg Denmark
- Department of Physical Therapy Marquette University Milwaukee WI USA
| | | | - Line B. Sørensen
- Center for Neuroplasticity and Pain (CNAP) Aalborg University Aalborg Denmark
| | - Hjalte H. Andersen
- Laboratory of Cutaneous Experimental Pain SMIAalborg University Aalborg Denmark
| | | |
Collapse
|
26
|
Zheng Z, Bai L, O'Loughlan M, Li CG, Xue CC. Does Electroacupuncture Have Different Effects on Peripheral and Central Sensitization in Humans: A Randomized Controlled Study. Front Integr Neurosci 2019; 13:61. [PMID: 31680888 PMCID: PMC6804574 DOI: 10.3389/fnint.2019.00061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/30/2019] [Indexed: 11/13/2022] Open
Abstract
Background Acupuncture is used to reduce chronic musculoskeletal pain. The common mechanism underlying these types of pain are peripheral and/or central sensitization. In the clinical setting, it is difficult to separate the peripheral from the central component of pain. Heat/capsaicin 45°C/0.075%-induced hyperalgesia provides a stable, human central sensitization model in which the peripheral component is also assessed. Aim This randomized, sham-controlled study aimed to investigate the effect of electroacupuncture (EA) on the severity of heat (peripheral sensitization) and mechanical hyperalgesia (central sensitization) in a heat/capsaicin pain model in humans. Methods Twenty-six healthy young participants (24 ± 3.9 years) were recruited. After baseline assessment, heat/capsaicin 45°C/0.075% was applied to the non-dominant forearm to induce hyperalgesia. The primary outcome measures were the size of the area of mechanical hyperalgesia, intensity of pain to heat stimulation and heat pain thresholds. The intensity of pain was recorded using modified 10-cm visual analogues scales (VAS). Participants were assessed at 70 min after the initial application of capsaicin then randomly allocated to receive either real electroacupuncture (REA, n = 14) or sham non-invasive EA (SEA, n = 12) for 30 min. The main outcome measures were assessed again immediately and then 90 min following EA. Credibility of blinding was assessed. Data were analyzed with t-tests or analysis of variance (ANOVA) where appropriate. Results After the model was established, the area of mechanical hyperalgesia was formed (55.64 cm2), as was heat hyperalgesia, as the rating to heat stimulation, increased from 2/10 to 6/10. The REA and SEA groups were comparable. Immediately after the allocated acupuncture treatment, the rating to heat stimulation was statistically significantly lower in the REA group (2.94 ± 1.64) than in the SEA group (4.62 ± 2.26) (p < 0.05). The area of mechanical hyperalgesia reduced significantly without any group difference. No group difference was detected in heat pain threshold. Blinding of the participants was successful. Conclusion Peripheral and central sensitization in the heat/capsaicin 45°C/0.075% model responded to EA differently, suggesting that acupuncture analgesia could vary, depending on the types of pain. This observation may explain some inconsistent findings from clinical trials of acupuncture.
Collapse
Affiliation(s)
- Zhen Zheng
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | | | | | - Chun Guang Li
- NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| | - Charlie C Xue
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| |
Collapse
|
27
|
Lötsch J, Walter C, Zunftmeister M, Zinn S, Wolters M, Ferreiros N, Rossmanith T, Oertel BG, Geisslinger G. A data science approach to the selection of most informative readouts of the human intradermal capsaicin pain model to assess pregabalin effects. Basic Clin Pharmacol Toxicol 2019; 126:318-331. [PMID: 31608551 DOI: 10.1111/bcpt.13337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/01/2019] [Indexed: 12/29/2022]
Abstract
Persistent and, in particular, neuropathic pain is a major healthcare problem with still insufficient pharmacological treatment options. This triggered research activities aimed at finding analgesics with a novel mechanism of action. Results of these efforts will need to pass through the phases of drug development, in which experimental human pain models are established components e.g. implemented as chemical hyperalgesia induced by capsaicin. We aimed at ranking the various readouts of a human capsaicin-based pain model with respect to the most relevant information about the effects of a potential reference analgesic. In a placebo-controlled, randomized cross-over study, seven different pain-related readouts were acquired in 16 healthy individuals before and after oral administration of 300 mg pregabalin. The sizes of the effect on pain induced by intradermal injection of capsaicin were quantified by calculating Cohen's d. While in four of the seven pain-related parameters, pregabalin provided a small effect judged by values of Cohen's d exceeding 0.2, an item categorization technique implemented as computed ABC analysis identified the pain intensities in the area of secondary hyperalgesia and of allodynia as the most suitable parameters to quantify the analgesic effects of pregabalin. Results of this study provide further support for the ability of the intradermal capsaicin pain model to show analgesic effects of pregabalin. Results can serve as a basis for the designs of studies where the inclusion of this particular pain model and pregabalin is planned.
Collapse
Affiliation(s)
- Jörn Lötsch
- Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany.,Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group Translational Medicine and Pharmacology, IME-TMP), Frankfurt am Main, Germany
| | - Carmen Walter
- Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group Translational Medicine and Pharmacology, IME-TMP), Frankfurt am Main, Germany
| | - Martin Zunftmeister
- Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany
| | - Sebastian Zinn
- Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany
| | - Miriam Wolters
- Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group Translational Medicine and Pharmacology, IME-TMP), Frankfurt am Main, Germany
| | - Nerea Ferreiros
- Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany
| | - Tanja Rossmanith
- Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group Translational Medicine and Pharmacology, IME-TMP), Frankfurt am Main, Germany
| | - Bruno G Oertel
- Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group Translational Medicine and Pharmacology, IME-TMP), Frankfurt am Main, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany.,Fraunhofer Institute of Molecular Biology and Applied Ecology-Project Group Translational Medicine and Pharmacology, IME-TMP), Frankfurt am Main, Germany
| |
Collapse
|
28
|
Szczot M, Liljencrantz J, Ghitani N, Barik A, Lam R, Thompson JH, Bharucha-Goebel D, Saade D, Necaise A, Donkervoort S, Foley AR, Gordon T, Case L, Bushnell MC, Bönnemann CG, Chesler AT. PIEZO2 mediates injury-induced tactile pain in mice and humans. Sci Transl Med 2019; 10:10/462/eaat9892. [PMID: 30305456 DOI: 10.1126/scitranslmed.aat9892] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/19/2018] [Indexed: 12/13/2022]
Abstract
Tissue injury and inflammation markedly alter touch perception, making normally innocuous sensations become intensely painful. Although this sensory distortion, known as tactile allodynia, is one of the most common types of pain, the mechanism by which gentle mechanical stimulation becomes unpleasant remains enigmatic. The stretch-gated ion channel PIEZO2 has been shown to mediate light touch, vibration detection, and proprioception. However, the role of this ion channel in nociception and pain has not been resolved. Here, we examined the importance of Piezo2 in the cellular representation of mechanosensation using in vivo imaging in mice. Piezo2-knockout neurons were completely insensitive to gentle dynamic touch but still responded robustly to noxious pinch. During inflammation and after injury, Piezo2 remained essential for detection of gentle mechanical stimuli. We hypothesized that loss of PIEZO2 might eliminate tactile allodynia in humans. Our results show that individuals with loss-of-function mutations in PIEZO2 completely failed to develop sensitization and painful reactions to touch after skin inflammation. These findings provide insight into the basis for tactile allodynia, identify the PIEZO2 mechanoreceptor as an essential mediator of touch under inflammatory conditions, and suggest that this ion channel might be targeted for treating tactile allodynia.
Collapse
Affiliation(s)
- Marcin Szczot
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jaquette Liljencrantz
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nima Ghitani
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Arnab Barik
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ruby Lam
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - James H Thompson
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana Bharucha-Goebel
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.,Department of Neurology, Children's National Medical Center, Washington, DC 20010, USA
| | - Dimah Saade
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Aaron Necaise
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sandra Donkervoort
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - A Reghan Foley
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Taylor Gordon
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laura Case
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - M Catherine Bushnell
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Carsten G Bönnemann
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander T Chesler
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
29
|
Neuroimaging of Pain: Human Evidence and Clinical Relevance of Central Nervous System Processes and Modulation. Anesthesiology 2019; 128:1241-1254. [PMID: 29494401 DOI: 10.1097/aln.0000000000002137] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuroimaging research has demonstrated definitive involvement of the central nervous system in the development, maintenance, and experience of chronic pain. Structural and functional neuroimaging has helped elucidate central nervous system contributors to chronic pain in humans. Neuroimaging of pain has provided a tool for increasing our understanding of how pharmacologic and psychologic therapies improve chronic pain. To date, findings from neuroimaging pain research have benefitted clinical practice by providing clinicians with an educational framework to discuss the biopsychosocial nature of pain with patients. Future advances in neuroimaging-based therapeutics (e.g., transcranial magnetic stimulation, real-time functional magnetic resonance imaging neurofeedback) may provide additional benefits for clinical practice. In the future, with standardization and validation, brain imaging could provide objective biomarkers of chronic pain, and guide treatment for personalized pain management. Similarly, brain-based biomarkers may provide an additional predictor of perioperative prognoses.
Collapse
|
30
|
Linde LD, Srbely JZ. The Acute Effect of Skin Preheating on Capsaicin-Induced Central Sensitization in Humans. Pain Pract 2019; 19:811-820. [PMID: 31231923 DOI: 10.1111/papr.12811] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 05/17/2019] [Accepted: 06/11/2019] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Topical capsaicin is commonly employed to experimentally induce central sensitization (CS) in humans. While previous studies have investigated the effect of skin preheating on the sensitizing effect of capsaicin, no studies have compared the synergistic effect of skin preheating on the magnitude of sensitization via topical capsaicin within the first 30 minutes of application. We tested the hypothesis that skin preheating potentiates the sensitizing effect of topical capsaicin by evoking a larger region of secondary hyperalgesia vs. topical capsaicin alone. METHODS Twenty young, healthy subjects each received topical capsaicin (Zostrix HP 0.075%) only (CAP), topical capsaicin with preheating (CAP + HEAT), and topical nonsensitizing placebo cream (CON) in a crossover design. Capsaicin and placebo creams were applied to a 50 cm2 area of the dorsal forearm. The CAP + HEAT session also included a 10-minute preheating session. Regions of secondary hyperalgesia were assessed using mechanical brush allodynia testing, and skin temperature was assessed via infrared thermography. Outcomes were normalized to baseline and compared at 10, 20, and 30 minutes after cream application. RESULTS The CAP + HEAT session led to a significantly larger area of secondary hyperalgesia compared to the CAP session as measured by brush allodynia (CON: 0 ± 0 cm; CAP: 2.08 ± 0.45 cm; CAP + HEAT: 3.70 ± 0.46 cm; P < 0.05) and skin temperature (CON: -2.92% ± 0.03%; CAP: -0.63% ± 0.09%; CAP + HEAT: 2.50% ± 0.11%; ( of baseline) P < 0.05). CONCLUSION Preheating amplifies the sensitizing effect of topical capsaicin within 30 minutes of application. The heat-capsaicin technique may be employed to assess differing magnitudes of CS induction and enables future studies investigating the development and progression of CS in humans.
Collapse
Affiliation(s)
- Lukas D Linde
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - John Z Srbely
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
31
|
Dowdle LT, Borckardt JJ, Back SE, Morgan K, Adams D, Madan A, Balliet W, Hanlon CA. Sensitized brain response to acute pain in patients using prescription opiates for chronic pain: A pilot study. Drug Alcohol Depend 2019; 200:6-13. [PMID: 31071496 PMCID: PMC6914256 DOI: 10.1016/j.drugalcdep.2019.02.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chronic opiate use leads to a sensitized behavioral response to acute pain, which in turn, leads to escalating doses of opiates. This study was designed to test the hypothesis that chronic opiate usage is also associated with a sensitized neurobiological response to acute pain in individuals that have used prescription opiates for 6 or more months. METHODS Fourteen patients with non-alcoholic chronic pancreatitis that have been taking prescription opiates for 6 or more months and 14 gender matched, non-opiate using controls were enrolled. Functional neuroimaging data was acquired while participants received blocks of thermal stimulation to their wrist (individually-tailored to their pain threshold). RESULTS Self-reported pain was significantly greater in opiate using patients (3.4 ± 3.4) than controls (0.2 ± 0.8: Brief Pain Inventory p < 0.005), however no significant difference between groups was observed in the individually-tailored pain thresholds. Opiate using patients evidenced a significantly greater response to pain than controls in two established nodes of the "Pain Matrix": somatosensory cortex (pFWE≤0.001) and anterior cingulate cortex (p ≤ 0.01). This response was positively correlated with prescribed morphine equivalent dosages (average: 133.5 ± 94.8 mg/day). CONCLUSION The findings suggest that in chronic pancreatitis patients, a dose of opiates that normalizes their behavioral response to acute pain is associated with an amplified neural response to acute pain. Further longitudinal studies are needed to determine if this neural sensitization hastens a behavioral tolerance to opiates or the development of an opioid use disorder.
Collapse
Affiliation(s)
- Logan T. Dowdle
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, USA,Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Jeffrey J. Borckardt
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, USA,Center for Biomedical Imaging, Medical University of South Carolina, Charleston, South Carolina, USA,Ralph H. Johnson VA Medical Center, Charleston, South Carolina, USA
| | - Sudie E. Back
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, USA,Ralph H. Johnson VA Medical Center, Charleston, South Carolina, USA
| | - Katherine Morgan
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - David Adams
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Alok Madan
- Houston Methodist Behavioral Health, Houston, Texas, USA
| | - Wendy Balliet
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Colleen A. Hanlon
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, South Carolina, USA,Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, USA,Center for Biomedical Imaging, Medical University of South Carolina, Charleston, South Carolina, USA,Ralph H. Johnson VA Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
32
|
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
|
33
|
Designing and conducting proof-of-concept chronic pain analgesic clinical trials. Pain Rep 2019; 4:e697. [PMID: 31583338 PMCID: PMC6749910 DOI: 10.1097/pr9.0000000000000697] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 09/24/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023] Open
Abstract
Introduction: The evolution of pain treatment is dependent on successful development and testing of interventions. Proof-of-concept (POC) studies bridge the gap between identification of a novel target and evaluation of the candidate intervention's efficacy within a pain model or the intended clinical pain population. Methods: This narrative review describes and evaluates clinical trial phases, specific POC pain trials, and approaches to patient profiling. Results: We describe common POC trial designs and their value and challenges, a mechanism-based approach, and statistical issues for consideration. Conclusion: Proof-of-concept trials provide initial evidence for target use in a specific population, the most appropriate dosing strategy, and duration of treatment. A significant goal in designing an informative and efficient POC study is to ensure that the study is safe and sufficiently sensitive to detect a preliminary efficacy signal (ie, a potentially valuable therapy). Proof-of-concept studies help avoid resources wasted on targets/molecules that are not likely to succeed. As such, the design of a successful POC trial requires careful consideration of the research objective, patient population, the particular intervention, and outcome(s) of interest. These trials provide the basis for future, larger-scale studies confirming efficacy, tolerability, side effects, and other associated risks.
Collapse
|
34
|
Machine-learned analysis of the association of next-generation sequencing-based human TRPV1 and TRPA1 genotypes with the sensitivity to heat stimuli and topically applied capsaicin. Pain 2019; 159:1366-1381. [PMID: 29596157 PMCID: PMC6012053 DOI: 10.1097/j.pain.0000000000001222] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Heat pain and its modulation by capsaicin varies among subjects in experimental and clinical settings. A plausible cause is a genetic component, of which TRPV1 ion channels, by their response to both heat and capsaicin, are primary candidates. However, TRPA1 channels can heterodimerize with TRPV1 channels and carry genetic variants reported to modulate heat pain sensitivity. To address the role of these candidate genes in capsaicin-induced hypersensitization to heat, pain thresholds acquired before and after topical application of capsaicin and TRPA1/TRPV1 exomic sequences derived by next-generation sequencing were assessed in n = 75 healthy volunteers and the genetic information comprised 278 loci. Gaussian mixture modeling indicated 2 phenotype groups with high or low capsaicin-induced hypersensitization to heat. Unsupervised machine learning implemented as swarm-based clustering hinted at differences in the genetic pattern between these phenotype groups. Several methods of supervised machine learning implemented as random forests, adaptive boosting, k-nearest neighbors, naive Bayes, support vector machines, and for comparison, binary logistic regression predicted the phenotype group association consistently better when based on the observed genotypes than when using a random permutation of the exomic sequences. Of note, TRPA1 variants were more important for correct phenotype group association than TRPV1 variants. This indicates a role of the TRPA1 and TRPV1 next-generation sequencing-based genetic pattern in the modulation of the individual response to heat-related pain phenotypes. When considering earlier evidence that topical capsaicin can induce neuropathy-like quantitative sensory testing patterns in healthy subjects, implications for future analgesic treatments with transient receptor potential inhibitors arise.
Collapse
|
35
|
Hansen MS, Becerra L, Dahl JB, Borsook D, Mårtensson J, Christensen A, Nybing JD, Havsteen I, Boesen M, Asghar MS. Brain resting-state connectivity in the development of secondary hyperalgesia in healthy men. Brain Struct Funct 2019; 224:1119-1139. [PMID: 30631932 DOI: 10.1007/s00429-018-01819-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 12/16/2018] [Indexed: 01/25/2023]
Abstract
Central sensitization is a condition in which there is an abnormal responsiveness to nociceptive stimuli. As such, the process may contribute to the development and maintenance of pain. Factors influencing the propensity for development of central sensitization have been a subject of intense debate and remain elusive. Injury-induced secondary hyperalgesia can be elicited by experimental pain models in humans, and is believed to be a result of central sensitization. Secondary hyperalgesia may thus reflect the individual level of central sensitization. The objective of this study was to investigate possible associations between increasing size of secondary hyperalgesia area and brain connectivity in known resting-state networks. We recruited 121 healthy participants (male, age 22, SD 3.35) who underwent resting-state functional magnetic resonance imaging. Prior to the scan session, areas of secondary hyperalgesia following brief thermal sensitization (3 min. 45 °C heat stimulation) were evaluated in all participants. 115 participants were included in the final analysis. We found a positive correlation (increasing connectivity) with increasing area of secondary hyperalgesia in the sensorimotor- and default mode networks. We also observed a negative correlation (decreasing connectivity) with increasing secondary hyperalgesia area in the sensorimotor-, fronto-parietal-, and default mode networks. Our findings indicate that increasing area of secondary hyperalgesia is associated with increasing and decreasing connectivity in multiple networks, suggesting that differences in the propensity for central sensitization, assessed as secondary hyperalgesia areas, may be expressed as differences in the resting-state central neuronal activity.
Collapse
Affiliation(s)
- Morten Sejer Hansen
- Department of Anaesthesiology, 4231, Centre of Head and Orthopaedics, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark.
- Department of Radiology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400, Copenhagen, NV, Denmark.
| | - Lino Becerra
- Invicro, A Konica Minolta Company, 27 Drydock Avenue, 7th Floor West, Boston, MA, 02210, USA
| | - Jørgen Berg Dahl
- Department of Anaesthesiology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400, Copenhagen, NV, Denmark
| | - David Borsook
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Psychiatry, McLean Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Johan Mårtensson
- Department of Clinical Sciences, Faculty of Medicine, Lund University, Box 213, 221 00, Lund, Sweden
| | - Anders Christensen
- Department of Radiology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400, Copenhagen, NV, Denmark
| | - Janus Damm Nybing
- Department of Radiology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400, Copenhagen, NV, Denmark
| | - Inger Havsteen
- Department of Radiology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Bispebjerg Bakke 23, 2400, Copenhagen, NV, Denmark
| | - Mikael Boesen
- Department of Radiology and the Parker Institute, Copenhagen University Hospital Bispebjerg and Frederiksberg, Bispebjerg Hospital, Bispebjerg Bakke 23, 2400, Copenhagen, NV, Denmark
| | - Mohammad Sohail Asghar
- Department of Neuroanaesthesiology, Neurocentre, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| |
Collapse
|
36
|
Street LM, Harris L, Curry RS, Eisenach JC. Capsaicin-induced pain and sensitisation in the postpartum period. Br J Anaesth 2018; 122:103-110. [PMID: 30579387 DOI: 10.1016/j.bja.2018.09.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 09/12/2018] [Accepted: 09/28/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recovery from Caesarean delivery in women and surgical nerve injury in animals after delivery is more rapid than expected, an effect reversed in animals by spinal injection of an oxytocin receptor antagonist. We hypothesised that endogenous modulation of acute pain is altered postpartum. METHODS Endogenous inhibition of acute pain in a conditioned pain modulation paradigm or endogenous sensitisation by topical capsaicin was tested in women who were breastfeeding 10-14 days after Caesarean delivery and age-matched controls (n=80 total: 20 per group and 20 per test). The study was powered to detect a difference in area of hyperalgesia after capsaicin of 33%. Capsaicin-evoked pain was recorded in women, and capsaicin-evoked mechanical hypersensitivity was measured in rats 48 h after delivery and in age-matched female and male animals. RESULTS There was no effect of the postpartum period in the endogenous sensitisation assay in women, and the conditioned pain modulation assay failed to produce analgesia in either group. Postpartum women, however, reported less intense pain than controls at the end of topical capsaicin exposure (1.3 [1.4] vs 2.0 [2.0] on 0-10 verbal scale), and acute hypersensitivity after capsaicin was less in postpartum than control rats (withdrawal threshold 25 [15] vs 3.6 [1] g). CONCLUSIONS These results agree with a recent report that oxytocin may desensitise the transient receptor potential for vanilloid-1 channel, although other explanations, including hormone effects, are possible. These results do not, however, support the inhibition of capsaicin-evoked spinal sensitisation in the postpartum period. CLINICAL TRIAL REGISTRATION NCT01843517.
Collapse
Affiliation(s)
- L M Street
- Department of Obstetrics and Gynecology, Augusta University, Augusta, GA, USA
| | - L Harris
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - R S Curry
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - J C Eisenach
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
37
|
|
38
|
Ferland CE, Villemure C, Michon PE, Gandhi W, Ma ML, Chouchou F, Parent AJ, Bushnell MC, Lavigne G, Rainville P, Ware MA, Jackson PL, Schweinhardt P, Marchand S. Multicenter assessment of quantitative sensory testing (QST) for the detection of neuropathic-like pain responses using the topical capsaicin model. Can J Pain 2018; 2:266-279. [PMID: 35005384 PMCID: PMC8730652 DOI: 10.1080/24740527.2018.1525682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background The use of quantitative sensory testing (QST) in multicenter studies has been quite limited, due in part to lack of standardized procedures among centers. Aim The aim of this study was to assess the application of the capsaicin pain model as a surrogate experimental human model of neuropathic pain in different centers and verify the variation in reports of QST measures across centers. Methods A multicenter study conducted by the Quebec Pain Research Network in six laboratories allowed the evaluation of nine QST parameters in 60 healthy subjects treated with topical capsaicin to model unilateral pain and allodynia. The same measurements (without capsaicin) were taken in 20 patients with chronic neuropathic pain recruited from an independent pain clinic. Results Results revealed that six parameters detected a significant difference between the capsaicin-treated and the control skin areas: (1) cold detection threshold (CDT) and (2) cold pain threshold (CPT) are lower on the capsaicin-treated side, indicating a decreased in cold sensitivity; (3) heat pain threshold (HPT) was lower on the capsaicin-treated side in healthy subjects, suggesting an increased heat pain sensitivity; (4) dynamic mechanical allodynia (DMA); (5) mechanical pain after two stimulations (MPS2); and (6) mechanical pain summation after ten stimulations (MPS10), are increased on the capsaicin-treated side, suggesting an increased in mechanical pain (P < 0.002). CDT, CPT and HPT showed comparable effects across all six centers, with CPT and HPT demonstrating the best sensitivity. Data from the patients showed significant difference between affected and unaffected body side but only with CDT. Conclusion These results provide further support for the application of QST in multicenter studies examining normal and pathological pain responses.
Collapse
Affiliation(s)
- Catherine E Ferland
- Quebec Pain Research Network, Université de Sherbrooke, Sherbrooke, QC, Canada.,Research Centre, Shriners Hospitals for Children-Canada, Montreal, QC, Canada.,Department of Anesthesia, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Chantal Villemure
- Alan Edwards Pain Management Unit, McGill University Health Centre, Montreal, QC, Canada
| | - Pierre-Emmanuel Michon
- Division des Neurosciences cliniques et cognitives, centre de recherche CERVO, Université Laval, Quebec, QC, Canada
| | - Wiebke Gandhi
- Centre for Integrative Neuroscience and Neurodynamics, School of Psychology and Clinical Language Sciences, University of Reading, Reading, UK
| | - My-Linh Ma
- Research Centre, Shriners Hospitals for Children-Canada, Montreal, QC, Canada
| | - Florian Chouchou
- Département santé buccale, Faculté de médecine dentaire, Université de Montréal, Montreal, QC, Canada
| | - Alexandre J Parent
- Quebec Pain Research Network, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - M Catherine Bushnell
- National Centre for Complementary and Integrative Health, NIH, Bethesda, MD, USA
| | - Gilles Lavigne
- Quebec Pain Research Network, Université de Sherbrooke, Sherbrooke, QC, Canada.,Département santé buccale, Faculté de médecine dentaire, Université de Montréal, Montreal, QC, Canada
| | - Pierre Rainville
- Quebec Pain Research Network, Université de Sherbrooke, Sherbrooke, QC, Canada.,Centre de recherche de l'Institut universitaire de gériatrie de Montréal (CRIUGM), Montreal, QC, Canada.,Département de stomatologie, Faculté de médecine dentaire, Université de Montréal, Montreal, QC, Canada
| | - Mark A Ware
- Quebec Pain Research Network, Université de Sherbrooke, Sherbrooke, QC, Canada.,Alan Edwards Pain Management Unit, McGill University Health Centre, Montreal, QC, Canada
| | - Philip L Jackson
- Quebec Pain Research Network, Université de Sherbrooke, Sherbrooke, QC, Canada.,Division des Neurosciences cliniques et cognitives, centre de recherche CERVO, Université Laval, Quebec, QC, Canada.,School of Psychology, Université Laval, Quebec, QC, Canada
| | - Petra Schweinhardt
- Quebec Pain Research Network, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Serge Marchand
- Quebec Pain Research Network, Université de Sherbrooke, Sherbrooke, QC, Canada.,Centre de recherche du CHUS, Sherbrooke, QC, Canada.,Department of Surgery, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Québec, Canada
| |
Collapse
|
39
|
Sauerstein K, Liebelt J, Namer B, Schmidt R, Rukwied R, Schmelz M. Low-Frequency Stimulation of Silent Nociceptors Induces Secondary Mechanical Hyperalgesia in Human Skin. Neuroscience 2018; 387:4-12. [DOI: 10.1016/j.neuroscience.2018.03.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 01/25/2018] [Accepted: 03/06/2018] [Indexed: 01/29/2023]
|
40
|
Huang YJ, Grau JW. Ionic plasticity and pain: The loss of descending serotonergic fibers after spinal cord injury transforms how GABA affects pain. Exp Neurol 2018; 306:105-116. [PMID: 29729247 PMCID: PMC5994379 DOI: 10.1016/j.expneurol.2018.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 04/24/2018] [Accepted: 05/01/2018] [Indexed: 12/14/2022]
Abstract
Activation of pain (nociceptive) fibers can sensitize neural circuits within the spinal cord, inducing an increase in excitability (central sensitization) that can foster chronic pain. The development of spinally-mediated central sensitization is regulated by descending fibers and GABAergic interneurons. In adult animals, the co-transporter KCC2 maintains a low intracellular concentration of the anion Cl-. As a result, when the GABA-A receptor is engaged, Cl- flows in the neuron which has a hyperpolarizing (inhibitory) effect. Spinal cord injury (SCI) can down-regulate KCC2 and reverse the flow of Cl-. Under these conditions, engaging the GABA-A receptor can have a depolarizing (excitatory) effect that fosters the development of nociceptive sensitization. The present paper explores how SCI alters GABA function and provides evidence that the loss of descending fibers alters pain transmission to the brain. Prior work has shown that, after SCI, administration of a GABA-A antagonist blocks the development of capsaicin-induced nociceptive sensitization, implying that GABA release plays an essential role. This excitatory effect is linked to serotonergic (5HT) fibers that descend through the dorsolateral funiculus (DLF) and impact spinal function via the 5HT-1A receptor. Supporting this, blocking the 5HT-1A receptor, or lesioning the DLF, emulated the effect of SCI. Conversely, spinal application of a 5HT-1A agonist up-regulated KCC2 and reversed the effect of bicuculline treatment. Finally, lesioning the DLF reversed how a GABA-A antagonist affects a capsaicin-induced aversion in a place conditioning task; in sham operated animals, bicuculline enhanced aversion whereas in DLF-lesioned rats biciculline had an antinociceptive effect.
Collapse
Affiliation(s)
- Yung-Jen Huang
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA.
| | - James W Grau
- Behavioral and Cellular Neuroscience, Department of Psychology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
41
|
Sjögren E, Kullenberg T, Jonzon B, Segerdahl M, Stålberg O, Halldin M, Sundgren-Andersson A. Clinical testing of three novel transient receptor potential cation channel subfamily V member 1 antagonists in a pharmacodynamic intradermal capsaicin model. Eur J Pain 2018; 22:1214-1228. [DOI: 10.1002/ejp.1209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2018] [Indexed: 11/11/2022]
Affiliation(s)
- E. Sjögren
- Department of Pharmacy; Uppsala University; Sweden
| | | | - B. Jonzon
- Medical Products Agency; Uppsala Sweden
| | - M. Segerdahl
- Lundbeck A/S, Valby, Denmark and Karolinska Institute; Stockholm Sweden
| | - O. Stålberg
- Division of Analytical Pharmaceutical Chemistry; Department of Medicinal Chemistry; Uppsala University; Sweden
| | - M.M. Halldin
- AlzeCure Foundation; Karolinska Institute Science Park; Huddinge Sweden
| | | |
Collapse
|
42
|
Characterization of a novel capsaicin/heat ongoing pain model. Eur J Pain 2017; 22:370-384. [DOI: 10.1002/ejp.1126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2017] [Indexed: 12/27/2022]
|
43
|
Randomized, double-blind, placebo-controlled, dose-escalation study: Investigation of the safety, pharmacokinetics, and antihyperalgesic activity of l-4-chlorokynurenine in healthy volunteers. Scand J Pain 2017; 17:243-251. [PMID: 29229209 DOI: 10.1016/j.sjpain.2017.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/16/2017] [Accepted: 05/17/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Neuropathic pain is a significant medical problem needing more effective treatments with fewer side effects. Overactive glutamatergic transmission via N-methyl-d-aspartate receptors (NMDARs) are known to play a role in central sensitization and neuropathic pain. Although ketamine, a NMDAR channel-blocking antagonist, is often used for neuropathic pain, its side-effect profile and abusive potential has prompted the search for a safer effective oral analgesic. A novel oral prodrug, AV-101 (l-4 chlorokynurenine), which, in the brain, is converted into one of the most potent and selective GlyB site antagonists of the NMDAR, has been demonstrated to be active in animal models of neuropathic pain. The two Phase 1 studies reported herein were designed to assess the safety and pharmacokinetics of AV-101, over a wide dose range, after daily dosing for 14-days. As secondary endpoints, AV-101 was evaluated in the capsaicin-induced pain model. METHODS The Phase 1A study was a single-site, randomized, double-blind, placebo-controlled, single oral ascending dose (30-1800mg) study involving 36 normal healthy volunteers. The Phase 1B study was a single-site randomized, double-blind, placebo-controlled, study of multiple ascending doses (360, 1080, and 1440mg/day) of AV-101 involving 50 normal healthy volunteers, to whom AV-101 or placebo were administered orally daily for 14 consecutive days. Subjects underwent PK blood analyses, laboratory assessments, physical examination, 12-lead ECG, ophthalmological examination, and various neurocognitive assessments. The effect of AV-101 was evaluated in the intradermally capsaicin-induced pain model (ClinicalTrials.gov Identifier: NCT01483846). RESULTS Two Phase 1, with an aggregate of 86 subjects, demonstrated that up to 14 days of oral AV-101, up to 1440mg per day, was safe and very well tolerated with AEs quantitively and qualitatively like those observed with placebo. Mean half-life values of AV-101 were consistent across doses, ranging with an average of 1.73h, with the highest Cmax (64.4μg/mL) and AUC0-t (196μgh/mL) values for AV-101 occurring in the 1440-mg dose group. In the capsaicin induce-pain model, there was no significant change in the area under the pain time curve (AUPC) for the spontaneous pain assessment between the treatment and the placebo groups on Day 1 or 14 (the primary endpoint). In contrast, there were consistent reductions at 60-180min on Day 1 after dosing for allodynia, mechanical hyperalgesia, heat hyperalgesia, and spontaneous pain, and on Day 14 after dosing for heat hyperalgesia. CONCLUSIONS Although, AV-101 did not reach statistical significance in reducing pain, there were consistent reductions, for allodynia pain and mechanical and heat hyperalgesia. Given the excellent safety profile and PK characteristics demonstrated by this study, future clinical trials of AV-101 in neuropathic pain are justified. IMPLICATIONS This article presents the safety and PK of AV-101, a novel oral prodrug producing a potent and selective GlyB site antagonist of the NMDA receptor. These data indicate that AV-101 has excellent safety and PK characteristics providing support for advancing AV-101 into Phase 2 studies in neuropathic pain, and even provides data suggesting that AV-101 may have a role in treating depression.
Collapse
|
44
|
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
|
45
|
Hansen MS, Wetterslev J, Pipper CB, Asghar MS, Dahl JB. Heat pain detection threshold is associated with the area of secondary hyperalgesia following brief thermal sensitization: a study of healthy male volunteers. J Pain Res 2017; 10:265-274. [PMID: 28184167 PMCID: PMC5291329 DOI: 10.2147/jpr.s121189] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Introduction The area of secondary hyperalgesia following brief thermal sensitization (BTS) of the skin and heat pain detection thresholds (HPDT) may both have predictive abilities in regards to pain sensitivity and clinical pain states. The association between HPDT and secondary hyperalgesia, however, remains unsettled, and the dissimilarities in physiologic properties suggest that they may represent 2 distinctively different pain entities. The aim of this study was to investigate the association between HPDT and BTS-induced secondary hyperalgesia. Methods A sample of 121 healthy male participants was included and tested on 2 separate study days with BTS (45°C, 3 minutes), HPDT, and pain during thermal stimulation (45°C, 1 minute). Areas of secondary hyperalgesia were quantified after monofilament pinprick stimulation. The pain catastrophizing scale (PCS) and hospital anxiety and depression scale (HADS) were also applied. Results A significant association between HPDT and the size of the area of secondary hyperalgesia (p<0.0001) was found. The expected change in area of secondary hyperalgesia due to a 1-degree increase in HPDT was estimated to be −27.38 cm2, 95% confidence interval (CI) of −37.77 to −16.98 cm2, with an R2 of 0.19. Likewise, a significant association between HADS-depression subscore and area of secondary hyperalgesia (p=0.046) was found, with an estimated expected change in secondary hyperalgesia to a 1-point increase in HADS-depression subscore of 11 cm2, 95% CI (0.19–21.82), and with R2 of 0.03. We found no significant associations between secondary hyperalgesia area and PCS score or pain during thermal stimulation. Conclusion HPDT and the area of secondary hyperalgesia after BTS are significantly associated; however, with an R2 of only 19%, HPDT only offers a modest explanation of the inter-participant variation in the size of the secondary hyperalgesia area elicited by BTS.
Collapse
Affiliation(s)
- Morten Sejer Hansen
- Department of Anesthesiology, 4231, Centre of Head and Orthopedics, Rigshospitalet
| | - Jørn Wetterslev
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Department 7812
| | | | | | - Jørgen Berg Dahl
- Department of Anesthesiology, Department Z, Bispebjerg Hospital, Copenhagen, Denmark
| |
Collapse
|
46
|
Förster M, Helfert S, Dierschke R, Großkopf M, Hüllemann P, Keller T, Baron R, Binder A. Evaluation of the antihyperalgesic effect of tapentadol in two human evoked pain models – the TapCapMentho pilot trial. Expert Opin Pharmacother 2016; 17:1717-25. [DOI: 10.1080/14656566.2016.1201071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- M. Förster
- Division of Neurological Pain Research and Therapy, Department of Neurology, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - S. Helfert
- Division of Neurological Pain Research and Therapy, Department of Neurology, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - R. Dierschke
- Division of Neurological Pain Research and Therapy, Department of Neurology, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - M. Großkopf
- Division of Neurological Pain Research and Therapy, Department of Neurology, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - P. Hüllemann
- Division of Neurological Pain Research and Therapy, Department of Neurology, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | | | - R. Baron
- Division of Neurological Pain Research and Therapy, Department of Neurology, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - A. Binder
- Division of Neurological Pain Research and Therapy, Department of Neurology, Christian-Albrechts-Universität Kiel, Kiel, Germany
| |
Collapse
|
47
|
Hansen MS, Asghar MS, Wetterslev J, Pipper CB, Johan Mårtensson J, Becerra L, Christensen A, Nybing JD, Havsteen I, Boesen M, Dahl JB. Is the Volume of the Caudate Nuclei Associated With Area of Secondary Hyperalgesia? - Protocol for a 3-Tesla MRI Study of Healthy Volunteers. JMIR Res Protoc 2016; 5:e117. [PMID: 27317630 PMCID: PMC4930528 DOI: 10.2196/resprot.5680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/08/2016] [Indexed: 11/13/2022] Open
Abstract
Background Experience and development of pain may be influenced by a number of physiological, psychological, and psychosocial factors. In a previous study we found differences in neuronal activation to noxious stimulation, and microstructural neuroanatomical differences, when comparing healthy volunteers with differences in size of the area of secondary hyperalgesia following a standardized burn injury. Objective We aim to investigate the degree of association between the volume of pain-relevant structures in the brain and the size of the area of secondary hyperalgesia following brief thermal sensitization. Methods The study consists of one experimental day, in which whole-brain magnetic resonance imaging (MRI) scans will be conducted including T1-weighed three-dimensional anatomy scan, diffusion tensor imaging, and resting state functional MRI. Before the experimental day, all included participants will undergo experimental pain testing in a parallel study (Clinicaltrials.gov Identifier: NCT02527395). Results from this experimental pain testing, as well as the size of the area of secondary hyperalgesia from the included participants, will be extracted from this parallel study. Results The association between the volume of pain-relevant structures in the brain and the area of secondary hyperalgesia will be investigated by linear regression of the estimated best linear unbiased predictors on the individual volumes of the pain relevant brain structures. Conclusions We plan to investigate the association between experimental pain testing parameters and the volume, connectivity, and resting state activity of pain-relevant structures in the brain. These results may improve our knowledge of the mechanisms responsible for the development of acute and chronic pain. ClinicalTrial Danish Research Ethics Committee (identifier: H-15010473). Danish Data Protection Agency (identifier: RH-2015-149). Clinicaltrials.gov NCT02567318; http://clinicaltrials.gov/ct2/show/NCT02567318 (Archived by WebCite at http://www.webcitation.org/6i4OtP0Oi)
Collapse
Affiliation(s)
- Morten Sejer Hansen
- Department of Anaesthesiology, 4231, Centre of head and orthopaedics, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Asad ABA, Seah S, Baumgartner R, Feng D, Jensen A, Manigbas E, Henry B, Houghton A, Evelhoch JL, Derbyshire SWG, Chin CL. Distinct BOLD fMRI Responses of Capsaicin-Induced Thermal Sensation Reveal Pain-Related Brain Activation in Nonhuman Primates. PLoS One 2016; 11:e0156805. [PMID: 27309348 PMCID: PMC4911046 DOI: 10.1371/journal.pone.0156805] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 05/18/2016] [Indexed: 01/05/2023] Open
Abstract
Background Approximately 20% of the adult population suffer from chronic pain that is not adequately treated by current therapies, highlighting a great need for improved treatment options. To develop effective analgesics, experimental human and animal models of pain are critical. Topically/intra-dermally applied capsaicin induces hyperalgesia and allodynia to thermal and tactile stimuli that mimics chronic pain and is a useful translation from preclinical research to clinical investigation. Many behavioral and self-report studies of pain have exploited the use of the capsaicin pain model, but objective biomarker correlates of the capsaicin augmented nociceptive response in nonhuman primates remains to be explored. Methodology Here we establish an aversive capsaicin-induced fMRI model using non-noxious heat stimuli in Cynomolgus monkeys (n = 8). BOLD fMRI data were collected during thermal challenge (ON:20 s/42°C; OFF:40 s/35°C, 4-cycle) at baseline and 30 min post-capsaicin (0.1 mg, topical, forearm) application. Tail withdrawal behavioral studies were also conducted in the same animals using 42°C or 48°C water bath pre- and post- capsaicin application (0.1 mg, subcutaneous, tail). Principal Findings Group comparisons between pre- and post-capsaicin application revealed significant BOLD signal increases in brain regions associated with the ‘pain matrix’, including somatosensory, frontal, and cingulate cortices, as well as the cerebellum (paired t-test, p<0.02, n = 8), while no significant change was found after the vehicle application. The tail withdrawal behavioral study demonstrated a significant main effect of temperature and a trend towards capsaicin induced reduction of latency at both temperatures. Conclusions These findings provide insights into the specific brain regions involved with aversive, ‘pain-like’, responses in a nonhuman primate model. Future studies may employ both behavioral and fMRI measures as translational biomarkers to gain deeper understanding of pain processing and evaluate the preclinical efficacy of novel analgesics.
Collapse
Affiliation(s)
- Abu Bakar Ali Asad
- Translational Biomarkers, Merck Research Laboratories, MSD, Singapore, Singapore
- * E-mail:
| | - Stephanie Seah
- Translational Biomarkers, Merck Research Laboratories, MSD, Singapore, Singapore
| | - Richard Baumgartner
- Biometrics Research, Biostatistics & Research Decision Sciences, Merck Research Laboratories, Merck & Co., Rahway, NJ, United States of America
| | - Dai Feng
- Biometrics Research, Biostatistics & Research Decision Sciences, Merck Research Laboratories, Merck & Co., Rahway, NJ, United States of America
| | - Andres Jensen
- Early Discovery Pharmacology, Merck Research Laboratories, MSD, Singapore, Singapore
| | | | - Brian Henry
- Early Discovery Pharmacology, Merck Research Laboratories, MSD, Singapore, Singapore
| | - Andrea Houghton
- Early Discovery Pharmacology, Merck Research Laboratories, Merck & Co., West Point, PA, United States of America
| | - Jeffrey L. Evelhoch
- Translational Biomarkers, Merck Research Laboratories, Merck & Co., West Point, PA, United States of America
| | - Stuart W. G. Derbyshire
- Dept of Psychology, National University of Singapore, Singapore, Singapore
- A*STAR-NUS Clinical Imaging Research Centre, Singapore, Singapore
| | - Chih-Liang Chin
- Translational Biomarkers, Merck Research Laboratories, MSD, Singapore, Singapore
| |
Collapse
|
49
|
High-Concentration L-Menthol Exhibits Counter-Irritancy to Neurogenic Inflammation, Thermal and Mechanical Hyperalgesia Caused by Trans-cinnamaldehyde. THE JOURNAL OF PAIN 2016; 17:919-29. [PMID: 27260636 DOI: 10.1016/j.jpain.2016.05.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/21/2016] [Accepted: 05/10/2016] [Indexed: 11/20/2022]
Abstract
UNLABELLED The transient receptor potential cation channel subfamily M 8 (TRPM8) agonist L-menthol has been used traditionally for its topical counterirritant properties. Although the use of topical L-menthol for pain is casuistically established, evidence regarding its efficacy is negligible. This study aimed to characterize the effect of L-menthol as a counterirritant on cutaneous pain and hyperalgesia provoked by topical application of the transient receptor potential cation channel, subfamily A, member 1 (TRPA1) agonist trans-cinnamaldehyde (CA). In a randomized, double-blinded study CA was applied to a 3 × 3-cm area of the volar forearm evoking neurogenic inflammation, pain, mechanical, and thermal hyperalgesia in 14 healthy volunteers. In different sessions, 10% CA alone or 40% L-menthol applied simultaneously with 10% CA were administered for 20 minutes, throughout which the subjects rated the pain intensity on a visual analogue scale of 0 to 10. Extensive quantitative sensory testing was conducted and superficial blood flow (neurogenic inflammation) was recorded. Administration of CA evoked spontaneous pain, neurogenic inflammation, thermal hyperalgesia, and primary and secondary mechanical hyperalgesia. Coadministration of topical L-menthol reduced spontaneous pain intensity (P < .01), neurogenic inflammation (P < .01), primary mechanical hyperalgesia (P < .05), secondary mechanical hyperalgesia (P < .05), and heat hyperalgesia (P < .05), but not cold hyperalgesia. L-menthol exhibited inhibitory effects on simultaneously established pain, hypersensitivity, and neurogenic inflammation in a human TRPA1-induced pain model. Potent TRPM8 agonists could be useful as topical antihyperalgesics. The study and the trial protocol is registered and approved by the local research ethics committee under the jurisdiction of the Danish Medicines Agency number N-20130005. The protocol also is registered at Clinicaltrials.gov under NCT02653703. PERSPECTIVE Drugs interacting with transient receptor potential channels are of great therapeutic potential. In the present study we established cutaneous pain and hyperalgesia using the TRPA1 agonist CA. Subsequently, we showed that the frequently used topical counterirritant and TRPM8 agonist, L-menthol, decreased evoked pain, hyperalgesia, and inflammation, indicating direct and indirect antinociceptive mechanisms.
Collapse
|
50
|
Hansen MS, Wetterslev J, Pipper CB, Asghar MS, Dahl JB. Is heat pain detection threshold associated with the area of secondary hyperalgesia following brief thermal sensitization? A study of healthy volunteers - design and detailed plan of analysis. BMC Anesthesiol 2016; 16:28. [PMID: 27246322 PMCID: PMC4888470 DOI: 10.1186/s12871-016-0193-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 05/15/2016] [Indexed: 12/12/2022] Open
Abstract
Background Several factors are believed to influence the development and experience of pain. Human clinical pain models are central tools, in the investigation of basic physiologic pain responses, and can be applied in patients as well as in healthy volunteers. Each clinical pain model investigates different aspects of the human pain response. Brief thermal sensitization induces a mild burn injury, resulting in development of primary hyperalgesia at the site of stimulation, and secondary hyperalgesia surrounding the site of stimulation. Central sensitization is believed to play an important role in the development of secondary hyperalgesia; however, a possible association of secondary hyperalgesia following brief thermal sensitization and other heat pain models remains unknown. Our aim with this study is to investigate how close the heat pain detection threshold is associated with the size of the area of secondary hyperalgesia induced by the clinical heat pain model: Brief thermal sensitization. Methods and design We aim to include 120 healthy participants. The participants will be tested on two separate study days with the following procedures: i) Brief thermal sensitization, ii) heat pain detection threshold and iii) pain during thermal stimulation. Additionally, the participants will be tested with the Pain Catastrophizing Scale and Hospital Anxiety and Depression Scale questionnaires. We conducted statistical simulations based on data from our previous study, to estimate an empirical power of 99.9 % with α of 0.05. We define that an R2 < 0.25 and predictive intervals larger than +/−150 cm2 are indications of a weak association. Discussion The area of secondary hyperalgesia may serve as a quantitative measure of the central sensitization induced by cutaneous heat stimulation, and thus may be a biomarker of an individual’s pain sensitivity. The number of studies investigating secondary hyperalgesia is growing; however basic knowledge of the physiologic aspects of secondary hyperalgesia in humans is still incomplete. We therefore find it interesting to investigate if HPDT, a known quantitative sensory test, is associated with areas of secondary hyperalgesia following brief thermal sensitization Trial registration Clinicaltrials.gov (Identifier: NCT02527395). Danish Research Ethics Committee (Identifier: H-8-2014-012). Danish Data Protection Agency (Identifier: 30-1436). Electronic supplementary material The online version of this article (doi:10.1186/s12871-016-0193-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Morten Sejer Hansen
- Department of Anaesthesiology, 4231, Centre of Head and Orthopaedics, Rigshospitalet, Blegdamsvej 9, Copenhagen, 2100, Denmark.
| | - Jørn Wetterslev
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, dep. 7812, Blegdamsvej 9, Copenhagen, 2100, Denmark
| | - Christian Bressen Pipper
- Section of Biostatistics, Faculty of Health, Copenhagen University, Øster Farigmagsgade 5, Copenhagen, 1014, Denmark
| | - Mohammad Sohail Asghar
- Department of Anaesthesiology, 4231, Centre of Head and Orthopaedics, Rigshospitalet, Blegdamsvej 9, Copenhagen, 2100, Denmark
| | - Jørgen Berg Dahl
- Department of Anaesthesiology, dep. Z, Bispebjerg Hospital, Bispebjerg Bakke 23, Copenhagen, 2400, Denmark
| |
Collapse
|