1
|
Alsaloum M, Dib-Hajj SD, Page DA, Ruben PC, Krainer AR, Waxman SG. Voltage-gated sodium channels in excitable cells as drug targets. Nat Rev Drug Discov 2025; 24:358-378. [PMID: 39901031 DOI: 10.1038/s41573-024-01108-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 02/05/2025]
Abstract
Excitable cells - including neurons, muscle cells and cardiac myocytes - are unique in expressing high densities of voltage-gated sodium (NaV) channels. This molecular adaptation enables these cells to produce action potentials, and is essential to their function. With the advent of the molecular revolution, the concept of 'the' sodium channel has been supplanted by understanding that excitable cells in mammals can express any of nine different forms of sodium channels (NaV1.1-NaV1.9). Selective expression in particular types of cells, together with a key role in controlling action potential firing, makes some of these NaV subtypes especially attractive molecular targets for drug development. Although these different channel subtypes display a common overall structure, differences in their amino acid sequences have provided a basis for the development of subtype-specific drugs. This approach has resulted in exciting progress in the development of drugs for epilepsy, cardiac disorders and pain. In this Review, we discuss recent progress in the development of drugs that selectively target each of the sodium channel subtypes.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Dana A Page
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Peter C Ruben
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | | | - Stephen G Waxman
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
2
|
Yang J, Xie YF, Smith R, Ratté S, Prescott SA. Discordance between preclinical and clinical testing of Na V 1.7-selective inhibitors for pain. Pain 2025; 166:481-501. [PMID: 39928833 PMCID: PMC11808711 DOI: 10.1097/j.pain.0000000000003425] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/19/2024] [Accepted: 08/13/2024] [Indexed: 10/26/2024]
Abstract
ABSTRACT The voltage-gated sodium channel Na V 1.7 plays an important role in pain processing according to genetic data. Those data made Na V 1.7 a popular drug target, especially since its relatively selective expression in nociceptors promised pain relief without the adverse effects associated with broader sodium channel blockade. Despite encouraging preclinical data in rodents, Na V 1.7-selective inhibitors have not yet proven effective in clinical trials. Discrepancies between preclinical and clinical results should raise alarms. We reviewed preclinical and clinical reports on the analgesic efficacy of Na V 1.7-selective inhibitors and found critical differences in several factors. Putting aside species differences, most preclinical studies tested young male rodents with limited genetic variability, inconsistent with the clinical population. Inflammatory pain was the most common preclinical chronic pain model whereas nearly all clinical trials focused on neuropathic pain despite some evidence suggesting Na V 1.7 channels are not essential for neuropathic pain. Preclinical studies almost exclusively measured evoked pain whereas most clinical trials assessed average pain intensity without distinguishing between evoked and spontaneous pain. Nearly all preclinical studies gave a single dose of drug unlike the repeat dosing used clinically, thus precluding preclinical data from demonstrating whether tolerance or other slow processes occur. In summary, preclinical testing of Na V 1.7-selective inhibitors aligned poorly with clinical testing. Beyond issues that have already garnered widespread attention in the pain literature, our results highlight the treatment regimen and choice of pain model as areas for improvement.
Collapse
Affiliation(s)
- Jane Yang
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yu-Feng Xie
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Russell Smith
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stéphanie Ratté
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Steven A. Prescott
- Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Devigili G, Lombardi R, Lauria G, Cazzato D. The Evolving Landscape of Small Fiber Neuropathy. Semin Neurol 2025; 45:132-144. [PMID: 39433284 DOI: 10.1055/s-0044-1791823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Small fiber neuropathy (SFN) belongs to a heterogeneous group of disorders in which thinly myelinated Aδ and unmyelinated C-fibers are primarily affected, leading to neuropathic pain and autonomic symptoms. SFN can be associated with systemic conditions such as diabetes, autoimmune diseases, exposure to drugs and toxins, and infection, with the list of associated diseases continuing to expand. Variants in the SCN9A, SCN10A, and SCN11A genes encoding Nav 1.7, Nav 1.8, and Nav 1.9 sodium channel subunits, as well as in the TRPA1 gene, have been found in SFN patients, expanding the spectrum of underlying conditions and enhancing our understanding of pathophysiological mechanisms. There is also growing interest in immune-mediated forms that could help identify potentially treatable subgroups. According to international criteria, diagnosis is established through clinical examination, the assessment of intraepidermal nerve fiber density, and/or quantitative sensory testing. Autonomic functional tests allow for a better characterization of dysautonomia in SFN, which can be subclinical. Other tests can support the diagnosis. Currently, the management of SFN prioritizes treating the underlying condition, if identified, within a multidisciplinary approach that combines symptomatic pain therapy, lifestyle changes, and biopsychological interventions. Emerging insights from the molecular characterization of SFN channelopathies hold promise for improving diagnosis, potentially leading to the discovery of new drugs and refining trial designs in the future. This article reviews the clinical presentation, diagnostic workup, and advancing knowledge of associated conditions and interventional management of SFN.
Collapse
Affiliation(s)
- Grazia Devigili
- Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy
| | - Raffaella Lombardi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy
| | - Giuseppe Lauria
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Daniele Cazzato
- Clinical Neurophysiology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta," Milan, Italy
| |
Collapse
|
4
|
Carmland ME, Kreutzfeldt MD, Holbech JV, Brask-Thomsen PK, Krøigård T, Hansen PN, Tankisi H, Jensen TS, Bach FW, Sindrup SH, Finnerup NB. The effect of lacosamide in peripheral neuropathic pain: A randomized, double-blind, placebo-controlled, phenotype-stratified trial. Eur J Pain 2024; 28:105-119. [PMID: 37565715 DOI: 10.1002/ejp.2165] [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: 05/10/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Neuropathic pain is common and difficult to treat. The sodium channel blocker lacosamide is efficacious in animal models of pain, but its effect on neuropathic pain in humans is inconclusive. METHODS In a multicentre, randomized, double-blinded placebo-controlled phenotype stratified trial, we examined if lacosamide produced better pain relief in patients with the irritable nociceptor phenotype compared to those without. The primary outcome was the change in daily average pain from baseline to last week of 12 weeks of treatment. Secondary and tertiary outcomes included pain relief, patient global impression of change and presence of 30% and 50% pain reduction. RESULTS The study was prematurely closed with 93 patients included and 63 randomized to lacosamide or placebo in a 2:1 ratio, of which 49 fulfilled the per protocol criteria and was used for the primary objective. We did not find a better effect of lacosamide in patients with the irritable nociceptor phenotype, the 95% CI for the primary objective was 0.41 (-1.2 to 2.0). For all patients randomized, lacosamide had no effect on the primary outcome, but significantly more patients were responders to lacosamide than during placebo, with an NNT of 4.0 (95% CI 2.3-16.1) and 5.0 (95% CI 2.8-24.5) for 30% and 50% pain reduction respectively. We did not identify any predictors for response. Lacosamide was generally well tolerated. CONCLUSION We could not confirm that lacosamide was more efficacious in patients with the irritable nociceptor type, but the study was prematurely closed, so we cannot exclude a small difference. SIGNIFICANCE Treatment of neuropathic pain is often a trial and error process. Little is known about which patient benefit from which kind of medication. The sodium channel blocker lacosamide shows variable effect on neuropathic pain. Pain sensory phenotype, as defined by quantitative sensory testing, did not predict response to treatment with lacosamide.
Collapse
Affiliation(s)
- Malin Erika Carmland
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | - Thomas Krøigård
- Department of Neurology, Odense University Hospital, Odense, Denmark
| | | | - Hatice Tankisi
- Department of Clinical Neurophysiology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Troels Staehelin Jensen
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Flemming Winther Bach
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Nanna Brix Finnerup
- Danish Pain Research Center, Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
5
|
Marchi M, Salvi E, Andelic M, Mehmeti E, D'Amato I, Cazzato D, Chiappori F, Lombardi R, Cartelli D, Devigili G, Dalla Bella E, Gerrits M, Almomani R, Malik RA, Ślęczkowska M, Mazzeo A, Gentile L, Dib-Hajj S, Waxman SG, Faber CG, Vecchio E, de Tommaso M, Lauria G. TRPA1 rare variants in chronic neuropathic and nociplastic pain patients. Pain 2023; 164:2048-2059. [PMID: 37079850 PMCID: PMC10443199 DOI: 10.1097/j.pain.0000000000002905] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/15/2022] [Accepted: 12/28/2022] [Indexed: 04/22/2023]
Abstract
Supplemental Digital Content is Available in the Text. TRPA1 gene is significantly enriched of rare variants in neuropathic pain and fibromyalgia patients, with itch or cold-induced pain as the most common features, opening new treatment opportunities. Missing aspects of the heritability of chronic neuropathic pain, as a complex adult-onset trait, may be hidden within rare variants with low effect on disease risk, unlikely to be resolved by a single-variant approach. To identify new risk genes, we performed a next-generation sequencing of 107 pain genes and collapsed the rare variants through gene-wise aggregation analysis. The optimal unified sequence kernel association test was applied to 169 patients with painful neuropathy, 223 patients with nociplastic pain (82 diagnosed with chronic widespread pain and 141 with fibromyalgia), and 216 healthy controls. Frequency and features of variants in TRPA1 , which was the most significant gene, were further validated in 2 independent cohorts of 140 patients with chronic pain (90 with painful neuropathy and 50 with chronic widespread pain) and 34 with painless neuropathy. The effect of aminoacidic changes were modeled in silico according to physicochemical characteristics. TRPA1 was significantly enriched of rare variants which significantly discriminated chronic pain patients from healthy controls after Bonferroni correction (P = 6.7 × 10−4, ρ = 1), giving a risk of 4.8-fold higher based on the simple burden test (P = 0.0015, OR = 4.8). Among the 32 patients harboring TRPA1 variants, 24 (75%) were diagnosed with nociplastic pain, either fibromyalgia (12; 37.5%) or chronic widespread pain (12; 37.5%), whereas 8 (25%) with painful neuropathy. Irrespective of the clinical diagnosis, 12 patients (38%) complained of itch and 10 (31.3%) of cold-induced or cold-accentuated pain, mostly episodic. Our study widens the spectrum of channelopathy-related chronic pain disorders and contributes to bridging the gap between phenotype and targeted therapies based on patients' molecular profile. 1_tzjjvsic Kaltura
Collapse
Affiliation(s)
- Margherita Marchi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Erika Salvi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Mirna Andelic
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Elkadia Mehmeti
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Ilaria D'Amato
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniele Cazzato
- Clinical Neurophysiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Federica Chiappori
- Consiglio Nazionale delle Ricerche, Istituto di Tecnologie Biomediche (CNR-ITB), Segrate (Milan), Italy
| | - Raffaella Lombardi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniele Cartelli
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Grazia Devigili
- Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Eleonora Dalla Bella
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Monique Gerrits
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Rowida Almomani
- Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Toxicogenomics, Maastricht University, Maastricht, the Netherlands
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Rayaz A. Malik
- Institute of Cardiovascular Sciences, Cardiac Centre, Faculty of Medical and Human Sciences, The University of Manchester and NIHR/WellcomeTrust Clinical Research Facility, Manchester, United Kingdom
- Research Division, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, Doha, Qatar
| | - Milena Ślęczkowska
- School of Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Toxicogenomics, Maastricht University, Maastricht, the Netherlands
| | - Anna Mazzeo
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Luca Gentile
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Stephen G. Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States
| | - Catharina G. Faber
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Eleonora Vecchio
- Neurophysiopathology Unit, DiBrain Department, Aldo Moro University, Bari, Italy
| | - Marina de Tommaso
- Neurophysiopathology Unit, DiBrain Department, Aldo Moro University, Bari, Italy
| | - Giuseppe Lauria
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
6
|
Andelic M, Salvi E, Marcuzzo S, Marchi M, Lombardi R, Cartelli D, Cazzato D, Mehmeti E, Gelemanovic A, Paolini M, Pardo C, D’Amato I, Hoeijmakers JGJ, Dib-Hajj S, Waxman SG, Faber CG, Lauria G. Integrative miRNA-mRNA profiling of human epidermis: unique signature of SCN9A painful neuropathy. Brain 2023; 146:3049-3062. [PMID: 36730021 PMCID: PMC10316770 DOI: 10.1093/brain/awad025] [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: 10/03/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 02/03/2023] Open
Abstract
Personalized management of neuropathic pain is an unmet clinical need due to heterogeneity of the underlying aetiologies, incompletely understood pathophysiological mechanisms and limited efficacy of existing treatments. Recent studies on microRNA in pain preclinical models have begun to yield insights into pain-related mechanisms, identifying nociception-related species differences and pinpointing potential drug candidates. With the aim of bridging the translational gap towards the clinic, we generated a human pain-related integrative miRNA and mRNA molecular profile of the epidermis, the tissue hosting small nerve fibres, in a deeply phenotyped cohort of patients with sodium channel-related painful neuropathy not responding to currently available therapies. We identified four miRNAs strongly discriminating patients from healthy individuals, confirming their effect on differentially expressed gene targets driving peripheral sensory transduction, transmission, modulation and post-transcriptional modifications, with strong effects on gene targets including NEDD4. We identified a complex epidermal miRNA-mRNA network based on tissue-specific experimental data suggesting a cross-talk between epidermal cells and axons in neuropathy pain. Using immunofluorescence assay and confocal microscopy, we observed that Nav1.7 signal intensity in keratinocytes strongly inversely correlated with NEDD4 expression that was downregulated by miR-30 family, suggesting post-transcriptional fine tuning of pain-related protein expression. Our targeted molecular profiling advances the understanding of specific neuropathic pain fine signatures and may accelerate process towards personalized medicine in patients with neuropathic pain.
Collapse
Affiliation(s)
- Mirna Andelic
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, 6229 ER Maastricht, The Netherlands
| | - Erika Salvi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Stefania Marcuzzo
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Margherita Marchi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Raffaella Lombardi
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Daniele Cartelli
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Daniele Cazzato
- Neurophysiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Elkadia Mehmeti
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Andrea Gelemanovic
- Biology of Robustness Group, Mediterranean Institute for Life Sciences (MedILS), 21000 Split, Croatia
| | - Matilde Paolini
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Carlotta Pardo
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Ilaria D’Amato
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
| | - Janneke G J Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, 6229 ER Maastricht, The Netherlands
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center+, 6229 ER Maastricht, The Netherlands
| | - Giuseppe Lauria
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy
| |
Collapse
|
7
|
Almomani R, Sopacua M, Marchi M, Ślęczkowska M, Lindsey P, de Greef BTA, Hoeijmakers JGJ, Salvi E, Merkies ISJ, Ferdousi M, Malik RA, Ziegler D, Derks KWJ, Boenhof G, Martinelli-Boneschi F, Cazzato D, Lombardi R, Dib-Hajj S, Waxman SG, Smeets HJM, Gerrits MM, Faber CG, Lauria G. Genetic Profiling of Sodium Channels in Diabetic Painful and Painless and Idiopathic Painful and Painless Neuropathies. Int J Mol Sci 2023; 24:ijms24098278. [PMID: 37175987 PMCID: PMC10179245 DOI: 10.3390/ijms24098278] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/15/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Neuropathic pain is a frequent feature of diabetic peripheral neuropathy (DPN) and small fiber neuropathy (SFN). Resolving the genetic architecture of these painful neuropathies will lead to better disease management strategies, counselling and intervention. Our aims were to profile ten sodium channel genes (SCG) expressed in a nociceptive pathway in painful and painless DPN and painful and painless SFN patients, and to provide a perspective for clinicians who assess patients with painful peripheral neuropathy. Between June 2014 and September 2016, 1125 patients with painful-DPN (n = 237), painless-DPN (n = 309), painful-SFN (n = 547) and painless-SFN (n = 32), recruited in four different centers, were analyzed for SCN3A, SCN7A-SCN11A and SCN1B-SCN4B variants by single molecule Molecular inversion probes-Next Generation Sequence. Patients were grouped based on phenotype and the presence of SCG variants. Screening of SCN3A, SCN7A-SCN11A, and SCN1B-SCN4B revealed 125 different (potential) pathogenic variants in 194 patients (17.2%, n = 194/1125). A potential pathogenic variant was present in 18.1% (n = 142/784) of painful neuropathy patients vs. 15.2% (n = 52/341) of painless neuropathy patients (17.3% (n = 41/237) for painful-DPN patients, 14.9% (n = 46/309) for painless-DPN patients, 18.5% (n = 101/547) for painful-SFN patients, and 18.8% (n = 6/32) for painless-SFN patients). Of the variants detected, 70% were in SCN7A, SCN9A, SCN10A and SCN11A. The frequency of SCN9A and SCN11A variants was the highest in painful-SFN patients, SCN7A variants in painful-DPN patients, and SCN10A variants in painless-DPN patients. Our findings suggest that rare SCG genetic variants may contribute to the development of painful neuropathy. Genetic profiling and SCG variant identification should aid in a better understanding of the genetic variability in patients with painful and painless neuropathy, and may lead to better risk stratification and the development of more targeted and personalized pain treatments.
Collapse
Affiliation(s)
- Rowida Almomani
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Maurice Sopacua
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Margherita Marchi
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Milena Ślęczkowska
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Patrick Lindsey
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Bianca T A de Greef
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Janneke G J Hoeijmakers
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Erika Salvi
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Ingemar S J Merkies
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
- Department of Neurology, Curaçao Medical Center, 4365+37Q, J. H. J. Hamelbergweg, Willemstad, Curacao
| | - Maryam Ferdousi
- Institute of Cardiovascular Sciences, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9P, UK
| | - Rayaz A Malik
- Institute of Cardiovascular Sciences, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9P, UK
- Weill Cornell Medicine-Qatar, Doha P.O. Box 24144, Qatar
| | - Dan Ziegler
- German Diabetes Centre, 40225 Düsseldorf, Germany
| | - Kasper W J Derks
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Gidon Boenhof
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, 40225 Düsseldorf, Germany
| | - Filippo Martinelli-Boneschi
- Laboratory of Human Genetics of Neurological Disorders, Institute of Experimental Neurology (INSPE), Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Daniele Cazzato
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Raffaella Lombardi
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Hubert J M Smeets
- Clinical Genomics Unit, Department of Genetics and Cell Biology, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department of Toxicogenomics, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Monique M Gerrits
- Department of Clinical Genetics, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Centre+, 6229 HX Maastricht, The Netherlands
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation "Carlo Besta" Neurological Institute, 20133 Milan, Italy
- Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan, 20157 Milan, Italy
| |
Collapse
|
8
|
Ovsepian SV, Waxman SG. Gene therapy for chronic pain: emerging opportunities in target-rich peripheral nociceptors. Nat Rev Neurosci 2023; 24:252-265. [PMID: 36658346 DOI: 10.1038/s41583-022-00673-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2022] [Indexed: 01/20/2023]
Abstract
With sweeping advances in precision delivery systems and manipulation of the genomes and transcriptomes of various cell types, medical biotechnology offers unprecedented selectivity for and control of a wide variety of biological processes, forging new opportunities for therapeutic interventions. This perspective summarizes state-of-the-art gene therapies enabled by recent innovations, with an emphasis on the expanding universe of molecular targets that govern the activity and function of primary sensory neurons and which might be exploited to effectively treat chronic pain.
Collapse
Affiliation(s)
- Saak V Ovsepian
- School of Science, Faculty of Engineering and Science, University of Greenwich London, Chatham Maritime, UK.
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA.
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
9
|
Themistocleous AC, Baskozos G, Blesneac I, Comini M, Megy K, Chong S, Deevi SVV, Ginsberg L, Gosal D, Hadden RDM, Horvath R, Mahdi-Rogers M, Manzur A, Mapeta R, Marshall A, Matthews E, McCarthy MI, Reilly MM, Renton T, Rice ASC, Vale TA, van Zuydam N, Walker SM, Woods CG, Bennett DLH. Investigating genotype-phenotype relationship of extreme neuropathic pain disorders in a UK national cohort. Brain Commun 2023; 5:fcad037. [PMID: 36895957 PMCID: PMC9991512 DOI: 10.1093/braincomms/fcad037] [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: 08/15/2022] [Revised: 10/12/2022] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
The aims of our study were to use whole genome sequencing in a cross-sectional cohort of patients to identify new variants in genes implicated in neuropathic pain, to determine the prevalence of known pathogenic variants and to understand the relationship between pathogenic variants and clinical presentation. Patients with extreme neuropathic pain phenotypes (both sensory loss and gain) were recruited from secondary care clinics in the UK and underwent whole genome sequencing as part of the National Institute for Health and Care Research Bioresource Rare Diseases project. A multidisciplinary team assessed the pathogenicity of rare variants in genes previously known to cause neuropathic pain disorders and exploratory analysis of research candidate genes was completed. Association testing for genes carrying rare variants was completed using the gene-wise approach of the combined burden and variance-component test SKAT-O. Patch clamp analysis was performed on transfected HEK293T cells for research candidate variants of genes encoding ion channels. The results include the following: (i) Medically actionable variants were found in 12% of study participants (205 recruited), including known pathogenic variants: SCN9A(ENST00000409672.1): c.2544T>C, p.Ile848Thr that causes inherited erythromelalgia, and SPTLC1(ENST00000262554.2):c.340T>G, p.Cys133Tr variant that causes hereditary sensory neuropathy type-1. (ii) Clinically relevant variants were most common in voltage-gated sodium channels (Nav). (iii) SCN9A(ENST00000409672.1):c.554G>A, pArg185His variant was more common in non-freezing cold injury participants than controls and causes a gain of function of NaV1.7 after cooling (the environmental trigger for non-freezing cold injury). (iv) Rare variant association testing showed a significant difference in distribution for genes NGF, KIF1A, SCN8A, TRPM8, KIF1A, TRPA1 and the regulatory regions of genes SCN11A, FLVCR1, KIF1A and SCN9A between European participants with neuropathic pain and controls. (v) The TRPA1(ENST00000262209.4):c.515C>T, p.Ala172Val variant identified in participants with episodic somatic pain disorder demonstrated gain-of-channel function to agonist stimulation. Whole genome sequencing identified clinically relevant variants in over 10% of participants with extreme neuropathic pain phenotypes. The majority of these variants were found in ion channels. Combining genetic analysis with functional validation can lead to a better understanding as to how rare variants in ion channels lead to sensory neuron hyper-excitability, and how cold, as an environmental trigger, interacts with the gain-of-function NaV1.7 p.Arg185His variant. Our findings highlight the role of ion channel variants in the pathogenesis of extreme neuropathic pain disorders, likely mediated through changes in sensory neuron excitability and interaction with environmental triggers.
Collapse
Affiliation(s)
| | - Georgios Baskozos
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Iulia Blesneac
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Maddalena Comini
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Karyn Megy
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Sam Chong
- National Hospital for Neurology and Neurosurgery, University College London Hospitals, London, UK
| | - Sri V V Deevi
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Lionel Ginsberg
- Department of Neurology, Royal Free Hospital, London, UK
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| | - David Gosal
- Salford Royal NHS Foundation Trust, Salford, UK
| | | | - Rita Horvath
- Wellcome Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | - Adnan Manzur
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Rutendo Mapeta
- NIHR BioResource, Cambridge University Hospitals NHS Foundation, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Andrew Marshall
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Division of Neuroscience and Experimental Psychology, University of Manchester, Manchester, UK
- Department of Clinical Neurophysiology, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Emma Matthews
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and the National Hospital of Neurology and Neurosurgery, London, UK
| | - Mark I McCarthy
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Mary M Reilly
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and the National Hospital of Neurology and Neurosurgery, London, UK
| | - Tara Renton
- King’s College Hospital NHS Foundation Trust, London, UK
| | - Andrew S C Rice
- Pain Research, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Pain Medicine, Chelsea and Westminster Hospital NHS Foundation Trust, London, UK
| | - Tom A Vale
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Natalie van Zuydam
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Suellen M Walker
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Christopher Geoffrey Woods
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - David L H Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
10
|
Maximizing treatment efficacy through patient stratification in neuropathic pain trials. Nat Rev Neurol 2023; 19:53-64. [PMID: 36400867 DOI: 10.1038/s41582-022-00741-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2022] [Indexed: 11/19/2022]
Abstract
Treatment of neuropathic pain remains inadequate despite the elucidation of multiple pathophysiological mechanisms and the development of promising therapeutic compounds. The lack of success in translating knowledge into clinical practice has discouraged pharmaceutical companies from investing in pain medicine; however, new patient stratification approaches could help bridge the translation gap and develop individualized therapeutic approaches. As we highlight in this article, subgrouping of patients according to sensory profiles and other baseline characteristics could aid the prediction of treatment success. Furthermore, novel outcome measures have been developed for patients with neuropathic pain. The extent to which sensory profiles and outcome measures can be employed in routine clinical practice and clinical trials and across distinct neuropathic pain aetiologies is yet to be determined. Improvements in animal models, drawing on our knowledge of human pain, and robust public-private partnerships will be needed to pave the way to innovative and effective pain medicine in the future.
Collapse
|
11
|
Dormer A, Narayanan M, Schentag J, Achinko D, Norman E, Kerrigan J, Jay G, Heydorn W. A Review of the Therapeutic Targeting of SCN9A and Nav1.7 for Pain Relief in Current Human Clinical Trials. J Pain Res 2023; 16:1487-1498. [PMID: 37168847 PMCID: PMC10166096 DOI: 10.2147/jpr.s388896] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/14/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction There is a great need to find alternative treatments for chronic pain which have become a healthcare problem. We discuss current therapeutic targeting Nav1.7. Areas Covered Nav1.7 is a sodium ion channel protein that is associated with several human pain genetic syndromes. It has been found that mutations associated with Nav1.7 lead to the loss of the ability to perceive pain in individuals that are otherwise normal. Several therapeutic interventions are presently undergoing preclinical and research using the methodology of damping Nav1.7 expressions as a methodology to decrease the sensation of pain leading to analgesia. Expert Opinion It is our strong belief that there is a viable future in the targeting of protein of Nav1.7 for the relief of chronic pain in humans. The review will look at the genomics associated with SCN1A and proteomic of Nav1.7 as a foundation to explain the mechanism of the therapeutic interventions targeting Nav1.7, the human disease that are associated with Nav1.7, and the current development of treatment for chronic pain whether in preclinical or clinical trials targeting Nav1.7 expressions. The development of therapeutic antagonists targeting Nav1.7 could be a viable alternative to the current treatments which have led to the opioid crisis. Therefore, Nav1.7 targeted treatment has a major clinical significance that will have positive consequences as it relates to chronic pain interventions.
Collapse
Affiliation(s)
- Anton Dormer
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
- Correspondence: Anton Dormer, Research and Development, PepVax, Inc, 8720 Georgia Ave #1000, Silver Spring, MD, 20910, USA, Email
| | | | - Jerome Schentag
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - Daniel Achinko
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - Elton Norman
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - James Kerrigan
- Research and Development, Navintus, Inc, Princeton, NJ, USA
| | - Gary Jay
- Research and Development, Navintus, Inc, Princeton, NJ, USA
| | | |
Collapse
|
12
|
Effects of Photodynamic Therapy on Nav1.7 Expression in Spinal Dorsal Root Ganglion Neurons. Curr Med Sci 2022; 42:1267-1272. [PMID: 36462133 DOI: 10.1007/s11596-022-2640-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/17/2022] [Indexed: 12/05/2022]
Abstract
OBJECTIVE The aim of this study was to examine the effects of photodynamic therapy (PDT) on the expression of Nav1.7 in spinal dorsal root ganglion (DRG) neurons. METHODS The primary DRG neurons from newborn SD rats were cultured. The cells were identified by neuron-specific enolase immunofluorescence staining. DRG neurons were divided into four groups: control group, photosensitizer group, laser group, and PDT group. The cell viability was detected by a cell counting kit-8 (CCK8) assay. qRT-PCR and Western blotting were used to determine the mRNA and protein expression levels of Nav1.7 in DRG neurons. RESULTS The purity of the cultured primary DRG neurons was greater than 90%. Compared with the control group, no significant change was found in the cell viability of the photosensitizer group, while the viability in the laser group and the PDT group was significantly reduced. The mRNA and protein expression levels of Nav1.7 were significantly greater in the laser group and the PDT group than in the control group. At the same time, the mRNA and protein expression levels of Nav1.7 were greater in the laser group than in the PDT group. CONCLUSION Both laser and PDT could upregulate the expression of Nav1.7 in DRG neurons, and the promoting effect might be related to the pain induced by clinical treatment. This study provides a research basis for the use of laser and PDT to treat pain. A better understanding of the relationship between Nav1.7 and PDT can help clinicians better manage PDT-related pain.
Collapse
|
13
|
Wu PM, Lin YC, Chiang CW, Cho HY, Chuang TH, Yu MC, Wu SN, Tu YF. Effective Modulation by Lacosamide on Cumulative Inhibition of INa during High-Frequency Stimulation and Recovery of INa Block during Conditioning Pulse Train. Int J Mol Sci 2022; 23:11966. [PMID: 36233266 PMCID: PMC9570249 DOI: 10.3390/ijms231911966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 12/03/2022] Open
Abstract
The effects of lacosamide (LCS, Vimpat®), an anti-convulsant and analgesic, on voltage-gated Na+ current (INa) were investigated. LCS suppressed both the peak (transient, INa(T)) and sustained (late, INa(L)) components of INa with the IC50 values of 78 and 34 μM found in GH3 cells and of 112 and 26 μM in Neuro-2a cells, respectively. In GH3 cells, the voltage-dependent hysteresis of persistent INa (INa(P)) during the triangular ramp pulse was strikingly attenuated, and the decaying time constant (τ) of INa(T) or INa(L) during a train of depolarizing pulses was further shortened by LCS. The recovery time course from the INa block elicited by the preceding conditioning train can be fitted by two exponential processes, while the single exponential increase in current recovery without a conditioning train was adequately fitted. The fast and slow τ's of recovery from the INa block by the same conditioning protocol arose in the presence of LCS. In Neuro-2a cells, the strength of the instantaneous window INa (INa(W)) during the rapid ramp pulse was reduced by LCS. This reduction could be reversed by tefluthrin. Moreover, LCS accelerated the inactivation time course of INa activated by pulse train stimulation, and veratridine reversed its decrease in the decaying τ value in current inactivation. The docking results predicted the capability of LCS binding to some amino-acid residues in sodium channels owing to the occurrence of hydrophobic contact. Overall, our findings unveiled that LCS can interact with the sodium channels to alter the magnitude, gating, voltage-dependent hysteresis behavior, and use dependence of INa in excitable cells.
Collapse
Affiliation(s)
- Po-Ming Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yu-Ching Lin
- Department of Physical Medicine and Rehabilitation, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Chi-Wu Chiang
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Hsin-Yen Cho
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Tzu-Hsien Chuang
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Meng-Cheng Yu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Fang Tu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| |
Collapse
|
14
|
Alcántara Montero A, Pacheco de Vasconcelos SR. Are we heading towards individualized neuropathic pain treatment? REVISTA ESPANOLA DE ANESTESIOLOGIA Y REANIMACION 2022; 69:510-511. [PMID: 36088266 DOI: 10.1016/j.redare.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/20/2021] [Indexed: 06/15/2023]
Affiliation(s)
- A Alcántara Montero
- Centro de Salud Manuel Encinas, Consultorio de Malpartida de Cáceres, Cáceres, Spain.
| | | |
Collapse
|
15
|
Strand N, Wie C, Peck J, Maita M, Singh N, Dumbroff J, Tieppo Francio V, Murphy M, Chang K, Dickerson DM, Maloney J. Small Fiber Neuropathy. Curr Pain Headache Rep 2022; 26:429-438. [PMID: 35384587 DOI: 10.1007/s11916-022-01044-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2022] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW This narrative review aims to summarize advances in the field of small fiber neuropathy made over the last decade, with emphasis on novel research highlighting the distinctive features of SFN. RECENT FINDINGS While the management of SFNs is ideally aimed at treating the underlying cause, most patients will require pain control via multiple, concurrent therapies. Herein, we highlight the most up-to-date information for diagnosis, medication management, interventional management, and novel therapies on the horizon. Despite the prevalence of small fiber neuropathies, there is no clear consensus on guidelines specific for the treatment of SFN. Despite the lack of specific guidelines for SFN treatment, the most recent general neuropathic pain guidelines are based on Cochrane studies and randomized controlled trials (RCTs) which have individually examined therapies used for the more commonly studied SFNs, such as painful diabetic neuropathy and HIV neuropathy. The recommendations from current guidelines are based on variables such as number needed to treat (NNT), safety, ease of use, and effect on quality of life.
Collapse
Affiliation(s)
- N Strand
- Division of Pain Medicine, Mayo Clinic Hospital, Phoenix, AZ, USA.
| | - C Wie
- Division of Pain Medicine, Mayo Clinic Hospital, Phoenix, AZ, USA
| | - J Peck
- Performing Arts Medicine Department, Shenandoah University, Winchester, USA
| | - M Maita
- Division of Pain Medicine, Mayo Clinic Hospital, Phoenix, AZ, USA
| | - N Singh
- OrthoAlabama Spine and Sports, Birmingham, AL, USA
| | - J Dumbroff
- Mount Sinai Morningside and West Department of Anesthesiology, New York, NY, USA
| | - V Tieppo Francio
- Department of Rehabilitation on Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - M Murphy
- Department of Rehabilitation on Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - K Chang
- Department of Anesthesiology and Critical Care, Emory University, Atlanta, GA, USA
| | - D M Dickerson
- NorthShore University HealthSystem, Evanston, IL, USA
- University of Chicago Medicine, Chicago,, IL, USA
| | - J Maloney
- Division of Pain Medicine, Mayo Clinic Hospital, Phoenix, AZ, USA
| |
Collapse
|
16
|
Alsaloum M, Waxman SG. iPSCs and DRGs: stepping stones to new pain therapies. Trends Mol Med 2022; 28:110-122. [PMID: 34933815 PMCID: PMC8810720 DOI: 10.1016/j.molmed.2021.11.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 02/03/2023]
Abstract
There is a pressing need for more effective nonaddictive treatment options for pain. Pain signals are transmitted from the periphery into the spinal cord via dorsal root ganglion (DRG) neurons, whose excitability is driven by voltage-gated sodium (NaV) channels. Three NaV channels (NaV1.7, NaV1.8, and NaV1.9), preferentially expressed in DRG neurons, play important roles in pain signaling in humans. Blockade of these channels may provide a novel approach to the treatment of pain, but clinical translation of preclinical results has been challenging, in part due to differences between rodent and human DRG neurons. Human DRG neurons and iPSC-derived sensory neurons (iPSC-SNs) provide new preclinical platforms that may facilitate the development of novel pain therapeutics.
Collapse
Affiliation(s)
- Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA; Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA; Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Center for Neuroscience & Regeneration Research, Yale University, West Haven, CT, USA; Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
17
|
Labau JIR, Alsaloum M, Estacion M, Tanaka B, Dib-Hajj FB, Lauria G, Smeets HJM, Faber CG, Dib-Hajj S, Waxman SG. Lacosamide Inhibition of Na V1.7 Channels Depends on its Interaction With the Voltage Sensor Domain and the Channel Pore. Front Pharmacol 2022; 12:791740. [PMID: 34992539 PMCID: PMC8724789 DOI: 10.3389/fphar.2021.791740] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022] Open
Abstract
Lacosamide, developed as an anti-epileptic drug, has been used for the treatment of pain. Unlike typical anticonvulsants and local anesthetics which enhance fast-inactivation and bind within the pore of sodium channels, lacosamide enhances slow-inactivation of these channels, suggesting different binding mechanisms and mode of action. It has been reported that lacosamide's effect on NaV1.5 is sensitive to a mutation in the local anesthetic binding site, and that it binds with slow kinetics to the fast-inactivated state of NaV1.7. We recently showed that the NaV1.7-W1538R mutation in the voltage-sensing domain 4 completely abolishes NaV1.7 inhibition by clinically-achievable concentration of lacosamide. Our molecular docking analysis suggests a role for W1538 and pore residues as high affinity binding sites for lacosamide. Aryl sulfonamide sodium channel blockers are also sensitive to substitutions of the W1538 residue but not of pore residues. To elucidate the mechanism by which lacosamide exerts its effects, we used voltage-clamp recordings and show that lacosamide requires an intact local anesthetic binding site to inhibit NaV1.7 channels. Additionally, the W1538R mutation does not abrogate local anesthetic lidocaine-induced blockade. We also show that the naturally occurring arginine in NaV1.3 (NaV1.3-R1560), which corresponds to NaV1.7-W1538R, is not sufficient to explain the resistance of NaV1.3 to clinically-relevant concentrations of lacosamide. However, the NaV1.7-W1538R mutation conferred sensitivity to the NaV1.3-selective aryl-sulfonamide blocker ICA-121431. Together, the W1538 residue and an intact local anesthetic site are required for lacosamide's block of NaV1.7 at a clinically-achievable concentration. Moreover, the contribution of W1538 to lacosamide inhibitory effects appears to be isoform-specific.
Collapse
Affiliation(s)
- Julie I R Labau
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States.,Department of Toxicogenomics, Clinical Genomics, Maastricht University Medical Centre+, Maastricht, Netherlands.,School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Matthew Alsaloum
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States.,Yale Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, United States.,Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT, United States
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Brian Tanaka
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Fadia B Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Giuseppe Lauria
- Neuroalgology Unit, IRCCS Foundation, "Carlo Besta" Neurological Institute, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Hubert J M Smeets
- Department of Toxicogenomics, Clinical Genomics, Maastricht University Medical Centre+, Maastricht, Netherlands.,School of Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Catharina G Faber
- Department of Neurology, School of Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, Netherlands
| | - Sulayman Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, United States.,Center for Neuroscience and Regeneration Research, Yale University, West Haven, CT, United States.,Rehabilitation Research Center, Veteran Affairs Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
18
|
Small-Fiber-Neuropathien. DGNEUROLOGIE 2022; 5. [PMCID: PMC9559077 DOI: 10.1007/s42451-022-00488-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Die Small-Fiber-Neuropathie (SFN) ist eine Erkrankung des peripheren Nervensystems aufgrund einer isolierten oder vorwiegenden Schädigung dünn myelinisierter Aδ-Fasern und/oder unmyelinisierter C‑Fasern. Für die sichere Diagnose einer SFN ist neben der klinischen Symptomatik mit Schmerzen und Sensibilitätsstörungen, typischerweise mit distal betonter Ausbreitung, der apparative Nachweis einer Rarefizierung oder einer Funktionsstörung der dünn myelinisierten Aδ-Fasern und/oder der unmyelinisierten C‑Fasern gefordert. Im vorliegenden Beitrag wird eine Übersicht über die diagnostischen Verfahren zum Nachweis einer SFN sowie über mögliche Ursachen und Behandlungsoptionen gegeben.
Collapse
|
19
|
Gossrau G, Sabatowski R. [Diagnostics and therapy of neuropathic pain]. Anaesthesist 2021; 70:993-1002. [PMID: 34676422 DOI: 10.1007/s00101-021-01039-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2021] [Indexed: 12/11/2022]
Abstract
Neuropathic pain is pain caused by a lesion or disease of the somatosensory nervous system. Scientific studies have shown that neuropathic pain is the result of complex altered signalling processes in the peripheral and central nervous system. Current forms of treatment of neuropathic pain are causally oriented but also aim at symptomatic analgesia by pharmacological and nonpharmacological methods. Furthermore, psychological pain management techniques are used in a supportive role. This review summarizes the contemporary diagnostics of neuropathic pain using frequent diseases as examples and presents the evidence from randomized controlled trials on the treatment of neuropathic pain. Treatment guidelines for pharmacological management of neuropathic pain include evidence-based use of antidepressants, anticonvulsants, opioids, capsaicin and lidocaine.
Collapse
Affiliation(s)
- G Gossrau
- Interdisziplinäres UniversitätsSchmerzCentrum, Universitätsklinikum Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.
| | - R Sabatowski
- Interdisziplinäres UniversitätsSchmerzCentrum, Universitätsklinikum Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland.,Klinik und Poliklinik für Anästhesiologie und Intensivmedizin, TU Dresden, Fetscherstr. 74, 01307, Dresden, Deutschland
| |
Collapse
|
20
|
Yuan JH, Estacion M, Mis MA, Tanaka BS, Schulman BR, Chen L, Liu S, Dib-Hajj FB, Dib-Hajj SD, Waxman SG. KCNQ variants and pain modulation: a missense variant in Kv7.3 contributes to pain resilience. Brain Commun 2021; 3:fcab212. [PMID: 34557669 PMCID: PMC8454204 DOI: 10.1093/braincomms/fcab212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/02/2022] Open
Abstract
There is a pressing need for understanding of factors that confer resilience to pain. Gain-of-function mutations in sodium channel Nav1.7 produce hyperexcitability of dorsal root ganglion neurons underlying inherited erythromelalgia, a human genetic model of neuropathic pain. While most individuals with erythromelalgia experience excruciating pain, occasional outliers report more moderate pain. These differences in pain profiles in blood-related erythromelalgia subjects carrying the same pain-causative Nav1.7 mutation and markedly different pain experience provide a unique opportunity to investigate potential genetic factors that contribute to inter-individual variability in pain. We studied a patient with inherited erythromelalgia and a Nav1.7 mutation (c.4345T>G, p. F1449V) with severe pain as is characteristic of most inherited erythromelalgia patients, and her mother who carries the same Nav1.7 mutation with a milder pain phenotype. Detailed six-week daily pain diaries of pain episodes confirmed their distinct pain profiles. Electrophysiological studies on subject-specific induced pluripotent stem cell-derived sensory neurons from each of these patients showed that the excitability of these cells paralleled their pain phenotype. Whole-exome sequencing identified a missense variant (c.2263C>T, p. D755N) in KCNQ3 (Kv7.3) in the pain resilient mother. Voltage-clamp recordings showed that co-expression of Kv7.2-wild type (WT)/Kv7.3-D755N channels produced larger M-currents than that of Kv7.2-WT/Kv7.3-WT. The difference in excitability of the patient-specific induced pluripotent stem cell-derived sensory neurons was mimicked by modulating M-current levels using the dynamic clamp and a model of the mutant Kv7.2-WT/Kv7.3-D755N channels. These results show that a 'pain-in-a-dish' model can be used to explicate genetic contributors to pain, and confirm that KCNQ variants can confer pain resilience via an effect on peripheral sensory neurons.
Collapse
Affiliation(s)
- Jun-Hui Yuan
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Mark Estacion
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Malgorzata A Mis
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Brian S Tanaka
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Betsy R Schulman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Lubin Chen
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Shujun Liu
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Fadia B Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Sulayman D Dib-Hajj
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT 06520, USA
- Center for Rehabilitation Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
21
|
Gemignani F, Bellanova MF, Saccani E, Pavesi G. Non-length-dependent small fiber neuropathy: Not a matter of stockings and gloves. Muscle Nerve 2021; 65:10-28. [PMID: 34374103 DOI: 10.1002/mus.27379] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/11/2021] [Accepted: 07/18/2021] [Indexed: 12/17/2022]
Abstract
The clinical spectrum of small fiber neuropathy (SFN) encompasses manifestations related to the involvement of thinly myelinated A-delta and unmyelinated C fibers, including not only the classical distal phenotype, but also a non-length-dependent (NLD) presentation that can be patchy, asymmetrical, upper limb-predominant, or diffuse. This narrative review is focused on NLD-SFN. The diagnosis of NLD-SFN can be problematic, due to its varied and often atypical presentation, and diagnostic criteria developed for distal SFN are not suitable for NLD-SFN. The topographic pattern of NLD-SFN is likely related to ganglionopathy restricted to the small neurons of dorsal root ganglia. It is often associated with systemic diseases, but about half the time is idiopathic. In comparison with distal SFN, immune-mediated diseases are more common than dysmetabolic conditions. Treatment is usually based on the management of neuropathic pain. Disease-modifying therapy, including immunotherapy, may be effective in patients with identified causes. Future research on NLD-SFN is expected to further clarify the interconnected aspects of phenotypic characterization, diagnostic criteria, and pathophysiology.
Collapse
Affiliation(s)
- Franco Gemignani
- Neurology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maria F Bellanova
- Laboratory of Neuromuscular Histopathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Elena Saccani
- Neurology Unit, Department of Specialized Medicine, University Hospital of Parma, Parma, Italy
| | - Giovanni Pavesi
- Neurology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Neuropathic pain remains difficult to treat. This review provides an update regarding recent advances in therapeutic management, particularly with regards to newer drugs, neurostimulation techniques and original study designs. RECENT FINDINGS Although the mainstay of neuropathic pain management is still represented by drug therapy, particularly antidepressants and antiepileptics, the place of nonpharmacological therapy including in particular brain neuromodulation techniques has substantially increased in recent years. Newer study designs are also increasingly implemented, based on in depth phenotypic profiling to achieve more individualized therapy, or on screening strategies to decrease placebo effect and contribute to increase assay sensitivity. These approaches are now considered the most promising to decrease therapeutic failures in neuropathic pain. SUMMARY Neuropathic pain management should not be restricted to pharmacotherapy but now encompasses multiple approaches including particularly neuromodulation techniques. Multimodal assessment can also help identify predictors of the response in clinical trials in order to ensure appropriate management.
Collapse
|
23
|
Pharmacotherapeutic Options for Managing Neuropathic Pain: A Systematic Review and Meta-Analysis. Pain Res Manag 2021; 2021:6656863. [PMID: 33986899 PMCID: PMC8093054 DOI: 10.1155/2021/6656863] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 04/20/2021] [Indexed: 01/02/2023]
Abstract
Despite an increasing number of available therapies, the treatment of neuropathic pain remains a major issue. Systematic reviews and meta-analyses indicate that only a minority of patients with neuropathic pain have an adequate response to pharmacological treatment and that most drugs have dose-limiting side effects. We conducted a systematic review and meta-analysis of randomised controlled trials published in the last five years. We searched for relevant papers within PubMed, EMBASE, the Cochrane Database of Systematic Reviews, and the Clinical Trials database (ClinicalTrials.gov). Two authors independently selected studies for inclusion, data extraction, and bias assessment. We identified 39 randomised controlled trials and included 16 in the meta-analysis. Trial outcomes were generally modest even for first-line drugs such as tricyclic antidepressants, serotonin-noradrenaline reuptake inhibitors, and gabapentinoids. Many drugs acting on new pain targets are currently under development. Clinical data are currently available for sodium channel isoform-specific antagonists, anti-nerve growth factor molecules, and fatty acid amide hydrolase inhibitors.
Collapse
|
24
|
Dickenson A. Why are sodium channel modulators not yet pharmacotherapeutic trailblazers for neuropathic pain? Expert Opin Pharmacother 2021; 22:1635-1637. [PMID: 33899639 DOI: 10.1080/14656566.2021.1917548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Anthony Dickenson
- Neuroscience, Physiology and Pharmacology, University College London, London, UK
| |
Collapse
|
25
|
Marshall A, Alam U, Themistocleous A, Calcutt N, Marshall A. Novel and Emerging Electrophysiological Biomarkers of Diabetic Neuropathy and Painful Diabetic Neuropathy. Clin Ther 2021; 43:1441-1456. [PMID: 33906790 DOI: 10.1016/j.clinthera.2021.03.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE Diabetic peripheral neuropathy (DPN) is the most common complication of diabetes. Small and large peripheral nerve fibers can be involved in DPN. Large nerve fiber damage causes paresthesia, sensory loss, and muscle weakness, and small nerve fiber damage is associated with pain, anesthesia, foot ulcer, and autonomic symptoms. Treatments for DPN and painful DPN (pDPN) pose considerable challenges due to the lack of effective therapies. To meet these challenges, there is a major need to develop biomarkers that can reliably diagnose and monitor progression of nerve damage and, for pDPN, facilitate personalized treatment based on underlying pain mechanisms. METHODS This study involved a comprehensive literature review, incorporating article searches in electronic databases (Google Scholar, PubMed, and OVID) and reference lists of relevant articles with the authors' substantial expertise in DPN. This review considered seminal and novel research and summarizes emerging biomarkers of DPN and pDPN that are based on neurophysiological methods. FINDINGS From the evidence gathered from 145 papers, this submission describes emerging clinical neurophysiological methods with potential to act as biomarkers for the diagnosis and monitoring of DPN as well as putative future roles as predictors of response to antineuropathic pain medication in pDPN. Nerve conduction studies only detect large fiber damage and do not capture pathology or dysfunction of small fibers. Because small nerve fiber damage is prominent in DPN, additional biomarkers of small nerve fiber function are needed. Activation of peripheral nociceptor fibers using laser, heat, or targeted electrical stimuli can generate pain-related evoked potentials, which are an objective neurophysiological measure of damage along the small fiber pathways. Assessment of nerve excitability, which provides a surrogate of axonal properties, may detect alterations in function before abnormalities are detected by nerve conduction studies. Microneurography and rate-dependent depression of the Hoffmann-reflex can be used to dissect underlying pain-generating mechanisms arising from the periphery and spinal cord, respectively. Their role in informing mechanistic-based treatment of pDPN as well as facilitating clinical trials design is discussed. IMPLICATIONS The neurophysiological methods discussed, although currently not practical for use in busy outpatient settings, detect small fiber and early large fiber damage in DPN as well as disclosing dominant pain mechanisms in pDPN. They are suited as diagnostic and predictive biomarkers as well as end points in mechanistic clinical trials of DPN and pDPN.
Collapse
Affiliation(s)
- Anne Marshall
- Musculoskeletal Biology, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom; Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Uazman Alam
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Andreas Themistocleous
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nigel Calcutt
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - Andrew Marshall
- Musculoskeletal Biology, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom; Department of Clinical Neurophysiology, The Walton Centre, Liverpool, United Kingdom; Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
26
|
Sachau J, Kersebaum D, Baron R, Dickenson AH. Unusual Pain Disorders - What Can Be Learned from Them? J Pain Res 2021; 13:3539-3554. [PMID: 33758536 PMCID: PMC7980038 DOI: 10.2147/jpr.s287603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Pain is common in many different disorders and leads to a significant reduction in quality of life in the affected patients. Current treatment options are limited and often result in insufficient pain relief, partly due to the incomplete understanding of the underlying pathophysiological mechanisms. The identification of these pathomechanisms is therefore a central object of current research. There are also a number of rare pain diseases, that are generally little known and often undiagnosed, but whose correct diagnosis and examination can help to improve the management of pain disorders in general. In some of these unusual pain disorders like sodium-channelopathies or sensory modulation disorder the underlying pathophysiological mechanisms have only recently been unravelled. These mechanisms might serve as pharmacological targets that may also play a role in subgroups of other, more common pain diseases. In other unusual pain disorders, the identification of pathomechanisms has already led to the development of new drugs. A completely new therapeutic approach, the gene silencing, can even stop progression in hereditary transthyretin amyloidosis and porphyria, ie in pain diseases that would otherwise be rapidly fatal if left untreated. Thus, pain therapists and researchers should be aware of these rare and unusual pain disorders as they offer the unique opportunity to study mechanisms, identify new druggable targets and finally because early diagnosis might save many patient lives.
Collapse
Affiliation(s)
- Juliane Sachau
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| | - Dilara Kersebaum
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| | - Ralf Baron
- Division of Neurological Pain Research and Therapy, Department of Neurology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, 24105, Germany
| | - Anthony H Dickenson
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, WC1E 6BT, UK
| |
Collapse
|
27
|
Hinckley CA, Kuryshev Y, Sers A, Barre A, Buisson B, Naik H, Hajos M. Characterization of Vixotrigine, a Broad-Spectrum Voltage-Gated Sodium Channel Blocker. Mol Pharmacol 2021; 99:49-59. [PMID: 33298520 DOI: 10.1124/molpharm.120.000079] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/15/2020] [Indexed: 02/02/2023] Open
Abstract
Voltage-gated sodium channels (Navs) are promising targets for analgesic and antiepileptic therapies. Although specificity between Nav subtypes may be desirable to target specific neural types, such as nociceptors in pain, many broadly acting Nav inhibitors are clinically beneficial in neuropathic pain and epilepsy. Here, we present the first systematic characterization of vixotrigine, a Nav blocker. Using recombinant systems, we find that vixotrigine potency is enhanced in a voltage- and use-dependent manner, consistent with a state-dependent block of Navs. Furthermore, we find that vixotrigine potently inhibits sodium currents produced by both peripheral and central nervous system Nav subtypes, with use-dependent IC50 values between 1.76 and 5.12 μM. Compared with carbamazepine, vixotrigine shows higher potency and more profound state-dependent inhibition but a similar broad spectrum of action distinct from Nav1.7- and Nav1.8-specific blockers. We find that vixotrigine rapidly inhibits Navs and prolongs recovery from the fast-inactivated state. In native rodent dorsal root ganglion sodium channels, we find that vixotrigine shifts steady-state inactivation curves. Based on these results, we conclude that vixotrigine is a broad-spectrum, state-dependent Nav blocker. SIGNIFICANCE STATEMENT: Vixotrigine blocks both peripheral and central voltage-gated sodium channel subtypes. Neurophysiological approaches in recombinant systems and sensory neurons suggest this block is state-dependent.
Collapse
Affiliation(s)
- Christopher A Hinckley
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Yuri Kuryshev
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Alissende Sers
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Alexander Barre
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Bruno Buisson
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Himanshu Naik
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| | - Mihaly Hajos
- Biogen, Cambridge, Massachusetts (C.A.H., H.N.); Charles River Laboratories Cleveland, Inc., Cleveland, Ohio (Y.K.); Neuroservice, Aix-en-Provence, France (A.S., A.B., B.B.); and Department of Comparative Medicine, Yale University, New Haven, Connecticut (M.H.)
| |
Collapse
|
28
|
Devigili G, Cazzato D, Lauria G. Clinical diagnosis and management of small fiber neuropathy: an update on best practice. Expert Rev Neurother 2020; 20:967-980. [PMID: 32654574 DOI: 10.1080/14737175.2020.1794825] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Small fiber neuropathy (SFN) is a heterogeneous group of disorders affecting thin myelinated Aδ and unmyelinated C fibers. Common symptoms include neuropathic pain and autonomic disturbances, and the typical clinical presentation is that of a length-dependent polyneuropathy, although other distributions could be present. AREA COVERED This review focuses on several aspects of SFN including etiology, clinical presentation, diagnostic criteria and tests, management, and future perspectives. Diagnostic challenges are discussed, encompassing the role of accurate and standardized assessment of symptoms and signs and providing clues for the clinical practice. The authors discuss the evidence in support of skin biopsy and quantitative sensory testing as diagnostic tests and present an overview of other diagnostic techniques to assess sensory and autonomic fibers dysfunction. The authors also suggest a systematic approach to the etiology including a set of laboratory tests and genetic examinations of sodium channelopathies and other rare conditions that might drive the therapeutic approach based on underlying cause or symptoms treatment. EXPERT OPINION SFN provides a useful model for neuropathic pain whose known mechanisms and cause could pave the way toward personalized treatments.
Collapse
Affiliation(s)
- Grazia Devigili
- Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta" , Milan, Italy
| | - Daniele Cazzato
- Neurophysiology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta" , Milan, Italy
| | - Giuseppe Lauria
- Neuroalgology Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta" , Milan, Italy.,Department of Biomedical and Clinical Sciences "Luigi Sacco", University of Milan , Milan, Italy
| |
Collapse
|
29
|
Painful and painless mutations of SCN9A and SCN11A voltage-gated sodium channels. Pflugers Arch 2020; 472:865-880. [PMID: 32601768 PMCID: PMC7351857 DOI: 10.1007/s00424-020-02419-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/25/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Abstract
Chronic pain is a global problem affecting up to 20% of the world’s population and has a significant economic, social and personal cost to society. Sensory neurons of the dorsal root ganglia (DRG) detect noxious stimuli and transmit this sensory information to regions of the central nervous system (CNS) where activity is perceived as pain. DRG neurons express multiple voltage-gated sodium channels that underlie their excitability. Research over the last 20 years has provided valuable insights into the critical roles that two channels, NaV1.7 and NaV1.9, play in pain signalling in man. Gain of function mutations in NaV1.7 cause painful conditions while loss of function mutations cause complete insensitivity to pain. Only gain of function mutations have been reported for NaV1.9. However, while most NaV1.9 mutations lead to painful conditions, a few are reported to cause insensitivity to pain. The critical roles these channels play in pain along with their low expression in the CNS and heart muscle suggest they are valid targets for novel analgesic drugs.
Collapse
|
30
|
Argyriou AA, Kalofonou F, Litsardopoulos P, Anastopoulou GG, Psimaras D, Bruna J, Kalofonos HP. Real world, open label experience with lacosamide against acute painful oxaliplatin-induced peripheral neurotoxicity. J Peripher Nerv Syst 2020; 25:178-183. [PMID: 32277545 DOI: 10.1111/jns.12374] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/11/2020] [Accepted: 04/05/2020] [Indexed: 12/19/2022]
Abstract
We report the outcome of a pilot, open-label study that tested the potential of lacosamide (200 mg/bi.d) as an effective and safe symptomatic treatment against acute painful oxaliplatin-induced peripheral neurotoxicity (OXAIPN). Lacosamide was introduced in 18 colorectal cancer patients with evidence of clinically significant acute, painful OXAIPN after infusion of the third course (T1) of oxaliplatin-based chemotherapy (FOLFOX4) and was maintained until completion of all 12 courses (T4). The OXA-Neuropathy Questionnaire (OXA-NQ) was used to record the severity of acute OXAIPN; the PI-NRS estimated the severity of neuropathic pain, while the chronic OXAIPN was graded with TNSc. The EuroQOL (EQ-5D) instrument was also applied. The Patient Global Impression of Change (PGIC) scale measured the lacosamide-attributed perception of change. LCM-responders were considered those with ≥50% reduction in PI-NRS and OXA-NQ scores at T4, compared to T1. Patients experienced on T1 a median number of acute OXAIPN symptoms of 4 and had a median neuropathic pain severity score of 6, which was strongly related to lower quality of life, according to EQ-VAS (P < .001). At T4, 12 patients (66.7%) were classified as responders. A significant clinical improvement was documented in the severity of acute OXAIPN and neuropathic pain in relation to lacosamide (P < .001) at T4 compared to T1, which was associated with improved EQ-VAS scores (P < .001). Twelve patients scored PGIC ≥5 (lacosamide-attributed) at T4. There were no incidences of early drop-outs for safety reasons. Lacosamide appears to be an effective and well-tolerated symptomatic treatment against acute, painful OXAIPN.
Collapse
Affiliation(s)
- Andreas A Argyriou
- Neurological Department, Saint Andrew's General Hospital of Patras, Patras, Greece.,Department of Medicine, Division of Oncology, Medical School, University of Patras, Patras, Greece
| | - Foteini Kalofonou
- Department of Oncology, Garry Weston Centre, Hammersmith Hospital, Imperial NHS Healthcare Trust, London, UK
| | | | | | - Dimitri Psimaras
- AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie Mazarin, Paris, France
| | - Jordi Bruna
- Neuro-Oncology Unit, Department of Neurology, Hospital Universitari de Bellvitge-ICO L'Hospitalet, IDIBELL, Barcelona, Spain
| | - Haralabos P Kalofonos
- Department of Medicine, Division of Oncology, Medical School, University of Patras, Patras, Greece
| |
Collapse
|