1
|
Kleppel DJ, Copeland R, Hussain N, Karri J, Wang E, D'Souza RS. Methodological and statistical characteristics of meta-analyses on spinal cord stimulation for chronic pain: a systematic review. Reg Anesth Pain Med 2025; 50:358-366. [PMID: 38388015 PMCID: PMC12015056 DOI: 10.1136/rapm-2023-105249] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/09/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND A growing number of meta-analyses (MA) have investigated the use of spinal cord stimulation (SCS) as a treatment modality for chronic pain. The quality of these MAs has not been assessed by validated appraisal tools. OBJECTIVE To examine the methodological characteristics and quality of MAs related to the use of SCS for chronic pain syndromes. EVIDENCE REVIEW An online literature search was conducted in Ovid MEDLINE(R), Ovid EMBASE, Ovid Cochrane Database of Systematic Reviews, and Scopus databases (January 1, 2000 through June 30, 2023) to identify MAs that investigated changes in pain intensity, opioid consumption, and/or physical function after SCS for the treatment of chronic pain. MA quality was assessed using A Measurement Tool to Assess Systematic Reviews (AMSTAR-2) critical appraisal tool. FINDINGS Twenty-five MAs were appraised in the final analysis. Three were considered "high" quality, three "low" quality, and 19 "critically low" quality, per the AMSTAR-2 criteria. There was no association between the publication year and AMSTAR-2 overall quality (β 0.043; 95% CI -0.008 to 0.095; p=0.097). There was an association between the impact factor and AMSTAR-2 overall quality (β 0.108; 95% CI 0.044 to 0.172; p=0.002), such that studies published in journals with higher impact factors were associated with higher overall quality. There was no association between the effect size and AMSTAR-2 overall quality (β -0.168; 95% CI -0.518 to 0.183; p=0.320).According to our power analysis, three studies were adequately powered (>80%) to reject the null hypothesis, while the remaining studies were underpowered (<80%). CONCLUSIONS The study demonstrates a critically low AMSTAR-2 quality for most MAs published on the use of SCS for treating chronic pain. Future MAs should improve study quality by implementing the AMSTAR-2 checklist items. PROSPERO REGISTRATION NUMBER CRD42023431155.
Collapse
Affiliation(s)
- Donald J Kleppel
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Royce Copeland
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, Texas, USA
| | - Nasir Hussain
- Department of Anesthesiology, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Jay Karri
- Departments of Orthopedic Surgery and Anesthesiology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Eric Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ryan S D'Souza
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
2
|
Widman AJ, Bashar T, Burton A, Clausen DM, Gupta P, Wolf DK, Folarin-Hines J, Payne M, Rogers JA, Meacham KW, Gereau RW, Gutruf P. Chronic, Battery-Free, Fully Implantable Multimodal Spinal Cord Stimulator for Pain Modulation in Small Animal Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415963. [PMID: 40184607 DOI: 10.1002/advs.202415963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/06/2025] [Indexed: 04/06/2025]
Abstract
Spinal cord stimulation (SCS) for chronic pain management is an invasive therapy involving surgical implantation of electrodes into spinal epidural space. While the clinical value and mechanistic action of the therapy is debated considerably in recent years, preclinical chronic studies employing rodent models can provide invaluable insights regarding the balance between efficacy and complications as well as mechanistic understanding of SCS therapy. However, current rodent compatible devices require tethered power delivery or bulky batteries, severely limiting the ability to probe long-term efficacy of SCS therapy. This work introduces a tether-free, small-footprint, fully implantable, battery-free SCS device compatible with rodent models, capable of delivering electrical stimulation to the spinal cord at a wide range of frequency, amplitude, and period via wireless communication adjustable on-demand without direct interaction with the animal. The presented device features capabilities of clinical SCS devices, with materials and processes amendable to scalable fabrication at a cost suitable for one-time use enabling high N studies. In this proof of concept, the implantable device serves to assess therapeutic efficacy of various clinically relevant SCS paradigms in alleviating neuropathic pain. This technology offers chronic stability and the potential to serve as the foundation for future research into the development of SCS therapeutic systems.
Collapse
Affiliation(s)
- Allie J Widman
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Taron Bashar
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, 85721, USA
| | - Alex Burton
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, 85721, USA
| | | | - Prashant Gupta
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Drew K Wolf
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Jakayla Folarin-Hines
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Maria Payne
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - John A Rogers
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Kathleen W Meacham
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Robert W Gereau
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, 63110, USA
- Department of Neuroscience, Washington University, St. Louis, MO, 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, 63110, USA
| | - Philipp Gutruf
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, 85721, USA
- Department of Electrical and Computer Engineering, University of Arizona, Tucson, AZ, 85721, USA
- Bio5 Institute, University of Arizona, Tucson, AZ, 85721, USA
- Neroscience GIDP, University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
3
|
Ramakrishna S, Moore M, Davies E, Merry AF, Sleigh J, Jowsey T. Long-term lived experiences of patients with chronic pain or angina pectoris treated with spinal cord stimulation: a qualitative study. BMJ Open 2025; 15:e082840. [PMID: 40132823 PMCID: PMC11934376 DOI: 10.1136/bmjopen-2023-082840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/23/2024] [Indexed: 03/27/2025] Open
Abstract
OBJECTIVE To explore the short-term and long-term lived experiences of patients with chronic pain and angina pectoris with spinal cord stimulation. DESIGN An interpretive qualitative study with thematic analysis of one-off, semistructured interviews, following Braun and Clarke (2006). SETTING A multidisciplinary, publicly funded pain service in Auckland, New Zealand. Patients usually undergo a comprehensive medical, psychological and functional assessment and an in-house pain management programme before proceeding to spinal cord stimulator implantation. PARTICIPANTS Participants implanted with a spinal cord stimulator between 1998 and 2019 who had their stimulator for ≥1 year, purposively sampled to increase the range of ethnicities. MAIN OUTCOME The themes identified from the interviews. RESULTS 24 participants with chronic pain of varied aetiology and a median (range) of 5.2 (2.4-23.2) years since stimulator implantation participated. 22 participants had the device in situ, and 2 had been explanted. Five main themes were identified: (1) embodiment: stimulator and body as one; (2) technical factors: batteries and type of stimulation; (3) improved well-being; (4) social connection and (5) healthcare system interaction. Most participants reported pain relief, but many had experienced complications and discomfort. They emphasised the importance of ongoing support from the pain service. Acceptance of pain, coping and embodiment emerged as common motifs across these themes. 21 participants were satisfied with their treatment. CONCLUSION Within the context of a multidisciplinary pain clinic, despite some discomfort and various complications, most participants valued the ongoing reduction of pain achieved with spinal cord stimulation. Timely access to support from the pain service influenced their experience and satisfaction with their stimulators. Acceptance of pain and embodiment of the stimulator helped participants adapt to living with their stimulator, often over many years.
Collapse
Affiliation(s)
- Shashikala Ramakrishna
- Department of Anaesthesiology, The University of Auckland Faculty of Medical and Health Sciences, School of Medicine, Auckland, New Zealand
| | - Matthew Moore
- Department of Anaesthesiology, The University of Auckland Faculty of Medical and Health Sciences, School of Medicine, Auckland, New Zealand
| | | | - Alan Forbes Merry
- Department of Anaesthesiology, The University of Auckland Faculty of Medical and Health Sciences, School of Medicine, Auckland, New Zealand
- Department of Cardiothoracic and ORL Anaesthesia, Auckland City Hospital, Auckland, New Zealand
| | - Jamie Sleigh
- Department of Anaesthesiology, The University of Auckland Faculty of Medical and Health Sciences, School of Medicine, Auckland, New Zealand
- Department of Anaesthesia, Waikato Hospital, Hamilton, New Zealand
| | - Tanisha Jowsey
- Department of Anaesthesiology, The University of Auckland Faculty of Medical and Health Sciences, School of Medicine, Auckland, New Zealand
- Bond University Faculty of Health Sciences and Medicine, Gold Coast, Queensland, Australia
| |
Collapse
|
4
|
Deshmukh A, Settell M, Cheng K, Knudsen B, Trevathan J, LaLuzerne M, Blanz S, Skubal A, Verma N, Romanauski B, Brucker-Hahn M, Lam D, Lavrov I, Suminski A, Weber D, Fisher L, Lempka S, Shoffstall A, Park H, Ross E, Zhang M, Ludwig K. Epidural spinal cord recordings (ESRs): sources of neural-appearing artifact in stimulation evoked compound action potentials. J Neural Eng 2025; 22:016050. [PMID: 39321832 DOI: 10.1088/1741-2552/ad7f8b] [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: 06/10/2024] [Accepted: 09/25/2024] [Indexed: 09/27/2024]
Abstract
Objective. Evoked compound action potentials (ECAPs) measured during epidural spinal cord stimulation (SCS) can help elucidate fundamental mechanisms for the treatment of pain and inform closed-loop control of SCS. Previous studies have used ECAPs to characterize neural responses to various neuromodulation therapies and have demonstrated that ECAPs are highly prone to multiple sources of artifact, including post-stimulus pulse capacitive artifact, electromyography (EMG) bleed-through, and motion artifact. However, a thorough characterization has yet to be performed for how these sources of artifact may contaminate recordings within the temporal window commonly used to determine activation of A-beta fibers in a large animal model.Approach. We characterized sources of artifacts that can contaminate the recording of ECAPs in an epidural SCS swine model using the Abbott Octrode™ lead.Main results. Spinal ECAP recordings can be contaminated by capacitive artifact, short latency EMG from nearby muscles of the back, and motion artifact. The capacitive artifact can appear nearly identical in duration and waveshape to evoked A-beta responses. EMG bleed-through can have phase shifts across the electrode array, similar to the phase shift anticipated by propagation of an evoked A-beta fiber response. The short latency EMG is often evident at currents similar to those needed to activate A-beta fibers associated with the treatment of pain. Changes in CSF between the cord and dura, and motion induced during breathing created a cyclic oscillation in all evoked components of recorded ECAPs.Significance. Controls must be implemented to separate neural signal from sources of artifact in SCS ECAPs. We suggest experimental procedures and reporting requirements necessary to disambiguate underlying neural response from these confounds. These data are important to better understand the framework for epidural spinal recordings (ESRs), with components such as ECAPs, EMG, and artifacts, and have important implications for closed-loop control algorithms to account for transient motion such as postural changes and cough.
Collapse
Affiliation(s)
- Ashlesha Deshmukh
- Wisconsin Institute for Translational Neuroengineering (WITNe), University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Megan Settell
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe), University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Kevin Cheng
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe), University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Bruce Knudsen
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe), University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States of America
| | - James Trevathan
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe), University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Maria LaLuzerne
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe), University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Stephan Blanz
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe), University of Wisconsin-Madison, Madison, WI, United States of America
| | - Aaron Skubal
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe), University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Nishant Verma
- Wisconsin Institute for Translational Neuroengineering (WITNe), University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States of America
- Abbott Neuromodulation, Plano, TX, United States of America
| | - Ben Romanauski
- Department of Neurology, Mayo Clinic, Rochester, MN, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America
| | - Meagan Brucker-Hahn
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - Danny Lam
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, United States of America
| | - Igor Lavrov
- Department of Neurology, Mayo Clinic, Rochester, MN, United States of America
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States of America
| | - Aaron Suminski
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe), University of Wisconsin-Madison, Madison, WI, United States of America
| | - Douglas Weber
- Mechanical Engineering Department, Carnegie Mellon University, Pittsburgh, PA, United States of America
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, United States of America
- NeuroMechatronics Lab, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Lee Fisher
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America
- Rehab Neural Engineering Laboratory (RNEL), University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Scott Lempka
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, United States of America
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States of America
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, United States of America
| | - Andrew Shoffstall
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
- Advanced Platform Technology Center, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, United States of America
| | - Hyunjoo Park
- Abbott Neuromodulation, Plano, TX, United States of America
| | - Erika Ross
- Abbott Neuromodulation, Plano, TX, United States of America
| | - Mingming Zhang
- Abbott Neuromodulation, Plano, TX, United States of America
| | - Kip Ludwig
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, United States of America
- Wisconsin Institute for Translational Neuroengineering (WITNe), University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States of America
- Department of Surgery, University of Wisconsin-Madison, Madison, WI, United States of America
| |
Collapse
|
5
|
Rigoard P, Ounajim A, Bouche B, Moens M, Goudman L, Eldabe S, Roulaud M, Lorgeoux B, Baron S, Nivole K, Many M, Lampert L, David R, Billot M. Comparison of Spinal Cord Stimulation, Dorsal Root Ganglion Stimulation, and Association of Both in Patients With Refractory Chronic Back and/or Lower Limb Neuropathic Pain: A Prospective, Randomized, Double-Blind, Cross-Over Trial (BOOST-DRG Study). Neuromodulation 2025; 28:283-296. [PMID: 39580743 DOI: 10.1016/j.neurom.2024.10.003] [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: 08/19/2024] [Revised: 09/06/2024] [Accepted: 10/06/2024] [Indexed: 11/26/2024]
Abstract
OBJECTIVES Spinal cord stimulation (SCS) and dorsal root ganglion stimulation (DRGS) have individually shown efficacy in relieving pain in patients with persistent spinal pain syndrome after spinal surgery (PSPS-T2). Combining SCS and DRGS simultaneously, along with Burst stimulation programming, may enhance the responder rate of patients with PSPS-T2. MATERIAL AND METHODS This study aimed to compare the pain relief (≥50%) responder rates in SCS, DRGS, and SCS+DGRS (DUAL) through a three-month randomized cross-over trial in patients with PSPS-T2. After the cross-over period, stimulation programming was switched to Burst. Secondary objectives included evaluating the clinical efficacy at three-, four-, six-, and 12-month follow-ups, assessing pain intensity, area of pain, area of paresthesia coverage, quality of life, functional disability, psychologic distress, medication intake, and the Multidimensional Clinical Response Index (MCRI). RESULTS The responder rate of pain relief was similar in SCS, DRGS, and DUAL (60%, p = 0.84) at the end of the cross-over period, increasing to 80% with the ability to switch between stimulation possibilities. Burst programming did not provide additional pain relief at the four-month follow-up (p = 0.99). Clinical outcomes significantly improved until 12-month follow-up compared with baseline. Considering a clinically significant increase of 1.05 of the MCRI, all patients were responders at three-, four-, and six- month follow-up, and 80% were responders at 12 months compared with baseline. CONCLUSIONS The full option to stimulate different neural structures, separately or simultaneously, led to improved responder rates, allowing patients to personalize treatment. A multidimensional assessment is essential to reveal the full potential benefits of neuromodulation in patients with chronic pain.
Collapse
Affiliation(s)
- Philippe Rigoard
- CHU de Poitiers, Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery Lab, F-86000 Poitiers, France; CHU de Poitiers, Department of Spine Neurosurgery and Neuromodulation, F-86000 Poitiers, France; Université de Poitiers, Prime Institute UPR, CNRS, ISAE-ENSMA, Poitiers, France
| | - Amine Ounajim
- CHU de Poitiers, Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery Lab, F-86000 Poitiers, France
| | - Bénédicte Bouche
- CHU de Poitiers, Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery Lab, F-86000 Poitiers, France; CHU de Poitiers, Department of Spine Neurosurgery and Neuromodulation, F-86000 Poitiers, France
| | - Maarten Moens
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Brussels, Belgium; STIMULUS consortium (reSearch and TeachIng neuroModULation Uz bruSsel), Vrije Universiteit Brussel, Brussels, Belgium; Department of Radiology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Lisa Goudman
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Brussels, Belgium; STIMULUS consortium (reSearch and TeachIng neuroModULation Uz bruSsel), Vrije Universiteit Brussel, Brussels, Belgium; Cluster Neurosciences, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium; Research Foundation-Flanders, Brussels, Belgium
| | - Sam Eldabe
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Manuel Roulaud
- CHU de Poitiers, Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery Lab, F-86000 Poitiers, France
| | - Bertille Lorgeoux
- CHU de Poitiers, Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery Lab, F-86000 Poitiers, France
| | - Sandrine Baron
- CHU de Poitiers, Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery Lab, F-86000 Poitiers, France
| | - Kévin Nivole
- CHU de Poitiers, Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery Lab, F-86000 Poitiers, France
| | - Mathilde Many
- CHU de Poitiers, Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery Lab, F-86000 Poitiers, France
| | - Lucie Lampert
- CHU de Poitiers, Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery Lab, F-86000 Poitiers, France
| | - Romain David
- CHU de Poitiers, Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery Lab, F-86000 Poitiers, France; CHU de Poitiers, Department of Physical Medicine and Rehabilitation, F-86000 Poitiers, France
| | - Maxime Billot
- CHU de Poitiers, Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery Lab, F-86000 Poitiers, France; Université de Poitiers and Université François Rabelais de Tours, Centre de Recherche sur la Cognition et l'Apprentissage, CNRS, F-86000 Poitiers, France.
| |
Collapse
|
6
|
Vanloon M, Van Broeckhoven T, Raymaekers V, De Ridder D, Billet B, Meeuws S, Menovsky T, Plazier M. Noninfectious Complications of Dorsal Root Ganglion Stimulation: A Systematic Review and Meta-Analysis. Neuromodulation 2025; 28:234-248. [PMID: 39601733 DOI: 10.1016/j.neurom.2024.10.010] [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: 09/04/2024] [Revised: 10/08/2024] [Accepted: 10/27/2024] [Indexed: 11/29/2024]
Abstract
INTRODUCTION Dorsal root ganglion stimulation (DRGS) has emerged as a promising treatment for chronic neuropathic pain. However, its safety and complications are not fully understood, with existing literature primarily based on case reports, observational studies, and data base analyses. This systematic review and meta-analysis aims to assess the prevalence of noninfectious complications associated with DRGS, focusing on the trial phase, postimplantation period, and revisions, while identifying risk factors for these outcomes. MATERIALS AND METHODS This systematic review adhered to Preferred Reporting Items for Systematic reviews and Meta-Analyses guidelines and was registered in the International Prospective Register of Systematic Reviews database. A comprehensive search was conducted across multiple data bases in June 2023. Studies included randomized and nonrandomized trials, and cohort studies involving ≥20 patients with DRGS. The exclusion criteria were studies that did not differentiate DRGS-specific complications, focused solely on infections, lacked sufficient data for prevalence estimation, or presented only subanalyses from larger studies. A meta-analysis of proportions was performed to estimate the overall prevalence of complications. RESULTS Thirteen studies with 634 participants were included. The pooled prevalence of all complications was 37% (95% CI: 19%-57%), with device-related complications being the most common at 27% (95% CI: 15%-42%). Lead fractures and migrations were the most frequently reported device-related complications with, respectively, 6% (95% CI: 2%-12%) and 6% (95% CI: 2%-10%). Procedure-related complications had a pooled prevalence of 1% (95% CI: 0%-5%), with dural puncture being the most common. The prevalence of DRGS explantations was 12%, primarily due to insufficient pain relief. CONCLUSIONS DRGS shows a safety profile comparable to that of spinal cord stimulation, with similar rates of lead migrations and fractures. Improvements in surgical techniques, technology, and clinician expertise are expected to reduce complications. Future research should standardize reporting practices and detail implantation techniques to better understand and refine best practices in DRGS implantation.
Collapse
Affiliation(s)
- Maarten Vanloon
- Faculty of Health, Medicine and Life Sciences, University Maastricht, Maastricht, The Netherlands; Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Medicine and Life Science, Hasselt University, Hasselt, Belgium.
| | - Tim Van Broeckhoven
- Faculty of Health, Medicine and Life Sciences, University Maastricht, Maastricht, The Netherlands
| | - Vincent Raymaekers
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Medicine and Life Science, Hasselt University, Hasselt, Belgium; Department of Neurosurgery, Antwerp University Hospital, Antwerp, Belgium
| | - Dirk De Ridder
- Department of Surgical Sciences, Section of Neurosurgery, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Bart Billet
- Department of Anesthesiology, AZ Delta, Roeselare, Belgium; STIMULUS research group, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sacha Meeuws
- Department of Neurosurgery Jessa Hospital, Hasselt, Belgium; Study and Training Centre Neurosurgery Virga Jesse, Hasselt, Belgium
| | - Tomas Menovsky
- Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium; Department of Neurosurgery, Antwerp University Hospital, Antwerp, Belgium
| | - Mark Plazier
- Faculty of Medicine and Life Science, Hasselt University, Hasselt, Belgium; Department of Neurosurgery Jessa Hospital, Hasselt, Belgium; Study and Training Centre Neurosurgery Virga Jesse, Hasselt, Belgium
| |
Collapse
|
7
|
Rigoard P, Ounajim A, Moens M, Goudman L, Roulaud M, Naiditch N, Boukenna R, Page P, Bouche B, Lorgeoux B, Baron S, Nivole K, Many M, Lampert L, Brumauld de Montgazon G, Roy-Moreau B, David R, Billot M. A digital tool for multidimensional assessment and prediction of treatment effectiveness in chronic pain management. iScience 2024; 27:111200. [PMID: 39758999 PMCID: PMC11700641 DOI: 10.1016/j.isci.2024.111200] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/22/2024] [Accepted: 10/15/2024] [Indexed: 01/07/2025] Open
Abstract
Given the multidimensional aspect of pain, the assessment of treatment efficacy is challenging. The prospective observational multicenter PREDIBACK study aimed to assess, compare, and predict the effectiveness of different treatments for persistent spinal pain syndrome type 2 (PSPS-T2) using a digital tool and the Multidimensional Clinical Response Index (MCRI) including pain intensity, functional disability, quality of life, anxiety and depression, and pain surface. Results indicated that neurostimulation was the most effective treatment at 3-, 6-, 9-, and 12-month follow-up compared to baseline, leading to significant improvements in pain, function, and quality of life, whereas optimized medical management (OMM) and spinal reoperation showed no significant benefits. Additionally, the study identified pain surface, BMI, and smoking status as predictors of treatment outcomes. These findings highlight the potential of digital medicine to improve patient care by providing data-driven insights and personalized treatment recommendations for PSPS-T2.
Collapse
Affiliation(s)
- Philippe Rigoard
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
- CHU de Poitiers, service de neurochirurgie du rachis, chirurgie de la douleur et du handicap, 86000 Poitiers, France
- Université de Poitiers, Pprime Institute UPR 3346, CNRS, ISAE-ENSMA, 86000 Poitiers, France
| | - Amine Ounajim
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
| | - Maarten Moens
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium
- STIMULUS consortium (reSearch and TeachIng neuroModULation Uz bruSsel), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Department of Radiology, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Lisa Goudman
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium
- STIMULUS consortium (reSearch and TeachIng neuroModULation Uz bruSsel), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
- Research Foundation—Flanders (FWO), 1090 Brussels, Belgium
| | - Manuel Roulaud
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
| | - Nicolas Naiditch
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
| | - Raouf Boukenna
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
| | - Philippe Page
- CHU de Poitiers, service de neurochirurgie du rachis, chirurgie de la douleur et du handicap, 86000 Poitiers, France
| | - Bénédicte Bouche
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
- CHU de Poitiers, service de neurochirurgie du rachis, chirurgie de la douleur et du handicap, 86000 Poitiers, France
| | - Bertille Lorgeoux
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
| | - Sandrine Baron
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
| | - Kevin Nivole
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
| | - Mathilde Many
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
| | - Lucie Lampert
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
| | | | - Brigitte Roy-Moreau
- Pain Evaluation and Treatment Centre, Nord Deux-Sèvres Hospital, 79350 Faye-l'Abbesse, France
| | - Romain David
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
- CHU de Poitiers, service de médecine physique et réadaptation, 86000 Poitiers, France
| | - Maxime Billot
- CHU de Poitiers, PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), 86000 Poitiers, France
- Centre de Recherche sur la Cognition et l’Apprentissage, Université de Poitiers, Université François Rabelais de Tours, CNRS, 86000 Poitiers, France
| |
Collapse
|
8
|
de Geus TJ, Franken G, Joosten EAJ. Spinal Cord Stimulation Paradigms and Alleviation of Neuropathic Pain Behavior in Experimental Painful Diabetic Polyneuropathy. Neuromodulation 2024; 27:1330-1337. [PMID: 39033461 DOI: 10.1016/j.neurom.2024.06.007] [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: 04/09/2024] [Revised: 06/01/2024] [Accepted: 06/20/2024] [Indexed: 07/23/2024]
Abstract
OBJECTIVES Spinal cord stimulation (SCS) is an alternative treatment option for painful diabetic polyneuropathy (PDPN). Differential target multiplexed (DTM)-SCS is proposed to be more effective than conventional (Con)-SCS. Animal studies are essential for understanding SCS mechanisms in PDPN pain relief. Although the Von Frey (VF) test is the gold standard for preclinical pain research, it has limitations. Operant testing using the conditioned place preference (CPP) test provides insights into spontaneous neuropathic pain relief and enhances the translatability of findings. This study aims to 1) use the CPP test to evaluate Con- and DTM-SCS effects on spontaneous neuropathic pain relief in PDPN animals and 2) investigate the correlation between mechanical hypersensitivity alleviation and spontaneous neuropathic pain relief. MATERIAL AND METHODS Diabetes was induced through streptozotocin injection in 32 rats; 16 animals developed PDPN and were implanted with a quadripolar lead. Rats were conditioned for Con-SCS (n = 8) or DTM-SCS (n = 7), and a preference score compared with sham was determined. After conditioning, a 30-minute SCS protocol was conducted. Mechanical sensitivity was assessed using VF before, during, and after SCS. RESULTS There were no significant chamber preference changes for DTM-SCS (p = 0.3449) or Con-SCS (p = 0.3632). Subgroups of responders and nonresponders were identified with significant increases in preference score for responders for both DTM-SCS (-266.6 to 119.8; p = 0.0238; n = 4) and Con-SCS (-350.7 to 88.46; p = 0.0148; n = 3). No strong correlation between SCS-induced spontaneous neuropathic pain relief and effects on mechanical hypersensitivity in PDPN animals is noted. CONCLUSIONS The CPP test is a valuable tool to test the efficacy of the pain-relieving potential of various SCS paradigms in PDPN animals. The results of this study show no differences in spontaneous neuropathic pain relief between DTM- and Con-SCS in PDPN animals. Furthermore, there is no correlation between the effect of SCS in spontaneous pain relief and hind paw mechanical hypersensitivity.
Collapse
Affiliation(s)
- Thomas J de Geus
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands.
| | - Glenn Franken
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Elbert A J Joosten
- Department of Anesthesiology and Pain Management, Maastricht University Medical Centre, Maastricht, The Netherlands; Mental Health and Neuroscience Research Institute, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
9
|
Miklós G, Halász L, Hasslberger M, Toth E, Manola L, Hagh Gooie S, van Elswijk G, Várkuti B, Erőss L. Sensory-substitution based sound perception using a spinal computer-brain interface. Sci Rep 2024; 14:24879. [PMID: 39438593 PMCID: PMC11496521 DOI: 10.1038/s41598-024-75779-2] [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: 08/19/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Sensory substitution offers a promising approach to restore lost sensory functions. Here we show that spinal cord stimulation (SCS), typically used for chronic pain management, can potentially serve as a novel auditory sensory substitution device. We recruited 13 patients undergoing SCS implantation and translated everyday sound samples into personalized SCS patterns during their trial phase. In a sound identification task-where chance-level performance was 33.3%-participants ( n = 8 ) achieved a mean accuracy of 72.8% using only SCS input. We observed a weak positive correlation between stimulation bitrate and identification accuracy. A follow-up discrimination task ( n = 5 ) confirmed that reduced bitrates significantly impaired participants' ability to distinguish between consecutive SCS patterns, indicating effective processing of additional information at higher bitrates. These findings demonstrate the feasibility of using existing SCS technology to create a novel neural interface for a sound prosthesis. Our results pave the way for future research to enhance stimulation fidelity, assess long-term training effects, and explore integration with other auditory aids for comprehensive hearing rehabilitation.
Collapse
Affiliation(s)
- Gabriella Miklós
- Institute of Neurosurgery and Neurointervention, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, Budapest, Hungary
- CereGate GmbH, München, Germany
| | - László Halász
- Institute of Neurosurgery and Neurointervention, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- Albert Szent-Györgyi Medical School, Doctoral School of Clinical Medicine, Clinical and Experimental Research for Reconstructive and Organ-Sparing Surgery, University of Szeged, Szeged, Hungary
| | | | | | | | | | | | | | - Loránd Erőss
- Institute of Neurosurgery and Neurointervention, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
10
|
D'Souza RS, Klasova J, Kleppel DJ, Prokop L, Hussain N. Hidden influence? Unmasking conflicts of interest from randomized clinical trials on spinal cord stimulation for chronic pain. Reg Anesth Pain Med 2024:rapm-2024-105903. [PMID: 39379095 DOI: 10.1136/rapm-2024-105903] [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: 07/26/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Randomized clinical trials (RCTs) are considered the gold standard for evaluating the efficacy of healthcare interventions. However, conflicts of interest (COIs) can compromise the scientific integrity in these trials. This study characterized COIs in RCTs on spinal cord stimulation for chronic pain, focusing on the prevalence, disclosure, and monetary value of COIs. METHODS This cross-sectional study analyzed RCTs published from January 1, 2013 to July 27, 2023. Primary outcomes included the presence, disclosure, and monetary value of COIs, while secondary outcomes assessed the presence of direct/indirect COIs, sponsor access to data, and associations between COIs and select variables, including journal impact factor, publication year, and study outcomes. RESULTS Of 38 RCTs, 30 (78.9%) reported COIs. On average, 35.6% of authors per RCT had at least one COI, with a mean of 0.7 COIs per author. The mean annual monetary value of COIs was US$41,157.83 per author per RCT. 29 RCTs (76.3%) had undisclosed COIs, with an average of 24.2% of authors per RCT having undisclosed COIs. Sponsor access to data was reported in 67.6% of RCTs. No associations were observed between the mean percentage of authors with COIs and the monetary value of COIs and select dependent variables (impact factor, publication year, and study outcomes). CONCLUSIONS A substantial majority of RCTs reported COIs with many authors having undisclosed conflicts, highlighting the need for stringent COI disclosure guidelines to maintain research integrity. Expanding COI registry systems globally and increasing non-industry funding are crucial steps toward enhancing transparency and reducing biases in medical research.
Collapse
Affiliation(s)
- Ryan S D'Souza
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Johana Klasova
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Donald J Kleppel
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Larry Prokop
- Mayo Clinic Libraries, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Nasir Hussain
- Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
11
|
Kallewaard JW, Duarte RV, Eldabe S, Thomson S. Comment on 'A sham-controlled, randomized trial of spinal cord stimulation for the treatment of pain in chronic pancreatitis' by Gulisano et al. Eur J Pain 2024; 28:1640-1641. [PMID: 39157998 DOI: 10.1002/ejp.4715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 08/20/2024]
Affiliation(s)
- Jan Willem Kallewaard
- Department of Anaesthesiology and Pain Management, Rijnstate Hospital, Elst, The Netherlands
- Department of Anesthesiology and Pain Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Rui V Duarte
- Saluda Medical Pty Ltd, Macquarie Park, New South Wales, Australia
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Sam Eldabe
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Simon Thomson
- Pain Medicine and Neuromodulation, Mid & South Essex University Hospitals, Essex, UK
| |
Collapse
|
12
|
Gulisano HA, Eriksen E, Bjarkam CR, Drewes AM, Olesen SS. A sham-controlled, randomized trial of spinal cord stimulation for the treatment of pain in chronic pancreatitis. Eur J Pain 2024; 28:1627-1639. [PMID: 38988274 DOI: 10.1002/ejp.2315] [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: 01/29/2024] [Revised: 05/30/2024] [Accepted: 06/30/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Spinal cord stimulation (SCS) has emerged as a treatment option for patients with chronic pancreatitis (CP) who experience pain that does not respond to standard interventions. However, there is a lack of sham-controlled trials to support its efficacy. METHODS This randomized, double-blinded, sham-controlled, cross-over trial enrolled 16 CP patients with insufficient pain relief from standard therapies. Patients underwent high-frequency (1000 Hz) paraesthesia-free SCS or sham for two 10-day stimulation periods, separated by a 3-day washout period. The primary outcome was daily pain intensity registered in a pain diary based on a numeric rating scale (NRS). Secondary outcomes included various questionnaires. Quantitative sensory testing was used to probe the pain system before and after interventions. RESULTS The average daily pain score on the NRS at baseline was 5.2 ± 1.9. After SCS, the pain score was 4.2 ± 2.1 compared to 4.3 ± 2.1 in the sham group (mean difference -0.1, 95% CI [-1.4 to 1.1]; P = 0.81). Similarly, no differences were observed between groups for the maximal daily pain score, secondary outcomes or quantitative sensory testing parameters. During an open-label, non-sham-controlled and non-blinded extension of the study, the average daily NRS was 5.2 ± 1.7 at baseline, 3.2 ± 1.8 at 3 months, 2.9 ± 1.9 at 6 months and 3.4 ± 2.2 at 12 months of follow-up (P = 0.001). CONCLUSION In this first sham-controlled trial of SCS in painful CP, we did not find evidence of short-term pain relief with paraesthesia-free high-frequency (1000 Hz) stimulation. However, evaluation of the long-term effect by larger sham-controlled trials with long-term follow-up is warranted. SIGNIFICANCE STATEMENT In this first sham-controlled trial to apply high-frequency (1000 Hz) spinal cord stimulation in patients with visceral pain due to chronic pancreatitis, we did not find evidence for clinically relevant pain relief. Taken together with potential procedure-related complications, adverse effects and costs associated with spinal cord stimulation, our findings question its use for management of visceral pain.
Collapse
Affiliation(s)
| | - Elin Eriksen
- Department of Neurosurgery, Aalborg University Hospital, Aalborg, Denmark
| | - Carsten Reidies Bjarkam
- Department of Neurosurgery, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University Hospital, Aalborg, Denmark
| | - Asbjørn Mohr Drewes
- Department of Clinical Medicine, Aalborg University Hospital, Aalborg, Denmark
- Centre for Pancreatic Diseases and Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Søren Schou Olesen
- Department of Clinical Medicine, Aalborg University Hospital, Aalborg, Denmark
- Centre for Pancreatic Diseases and Mech-Sense, Department of Gastroenterology and Hepatology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
13
|
Vishnevetsky A, Romanow G, Levy M. A transcutaneous electrical nerve stimulation device for the relief of neuropathic pain in NMOSD: A randomized, double-blind, sham-controlled trial. Mult Scler J Exp Transl Clin 2024; 10:20552173241301018. [PMID: 39651334 PMCID: PMC11622348 DOI: 10.1177/20552173241301018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/28/2024] [Indexed: 12/11/2024] Open
Abstract
Background Individuals with neuromyelitis optica spectrum disorder (NMOSD) often suffer from severe, disabling, and treatment-refractory neuropathic pain. Transcutaneous electrical nerve stimulation (TENS) therapy is a non-invasive, pain-modifying device. Objective To determine whether TENS therapy is safe, tolerable, and effective for neuropathic pain in patients with NMOSD. Methods We conducted a four-week, randomized, double-blind, sham-controlled, remote trial of TENS in patients with NMOSD who have neuropathic pain, followed by a 12-week open-label extension period. The difference in the Numeric Rating Scale current pain scores between 0 weeks and 4 weeks was the primary outcome measure. Results Forty-six patients (23 per arm) were enrolled in this trial, of which 40 were included in the primary analysis (four in the intervention arm and two in the sham arm withdrew prior to assessment of the primary outcome). Both the sham and intervention arms demonstrated significant decreases in average pain, worst pain, and current pain rating between baseline and 4 weeks, but there was no significant difference between the two arms. Conclusions In conclusion, there was no demonstrated benefit of TENS over sham TENS treatment, however, both arms demonstrated significant decreases in reported pain between baseline and 4 weeks. This trial is registered with ClinicalTrials.gov, NCT04614454.
Collapse
Affiliation(s)
- Anastasia Vishnevetsky
- Division of Neuroimmunology & Neuroinfectious Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gabriela Romanow
- Division of Neuroimmunology & Neuroinfectious Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael Levy
- Division of Neuroimmunology & Neuroinfectious Disease, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Røikjer J, Borbjerg MK, Andresen T, Giordano R, Hviid CVB, Mørch CD, Karlsson P, Klonoff DC, Arendt-Nielsen L, Ejskjaer N. Diabetic Peripheral Neuropathy: Emerging Treatments of Neuropathic Pain and Novel Diagnostic Methods. J Diabetes Sci Technol 2024:19322968241279553. [PMID: 39282925 PMCID: PMC11571639 DOI: 10.1177/19322968241279553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
BACKGROUND Diabetic peripheral neuropathy (DPN) is a prevalent and debilitating complication of diabetes, often leading to severe neuropathic pain. Although other diabetes-related complications have witnessed a surge of emerging treatments in recent years, DPN has seen minimal progression. This stagnation stems from various factors, including insensitive diagnostic methods and inadequate treatment options for neuropathic pain. METHODS In this comprehensive review, we highlight promising novel diagnostic techniques for assessing DPN, elucidating their development, strengths, and limitations, and assessing their potential as future reliable clinical biomarkers and endpoints. In addition, we delve into the most promising emerging pharmacological and mechanistic treatments for managing neuropathic pain, an area currently characterized by inadequate pain relief and a notable burden of side effects. RESULTS Skin biopsies, corneal confocal microscopy, transcutaneous electrical stimulation, blood-derived biomarkers, and multi-omics emerge as some of the most promising new techniques, while low-dose naltrexone, selective sodium-channel blockers, calcitonin gene-related peptide antibodies, and angiotensin type 2 receptor antagonists emerge as some of the most promising new drug candidates. CONCLUSION Our review concludes that although several promising diagnostic modalities and emerging treatments exist, an ongoing need persists for the further development of sensitive diagnostic tools and mechanism-based, personalized treatment approaches.
Collapse
Affiliation(s)
- Johan Røikjer
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
- Integrative Neuroscience, Aalborg University, Aalborg, Denmark
- Department Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Endocrinology, Aalborg University Hospital, Aalborg, Denmark
| | - Mette Krabsmark Borbjerg
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
- Integrative Neuroscience, Aalborg University, Aalborg, Denmark
| | - Trine Andresen
- Integrative Neuroscience, Aalborg University, Aalborg, Denmark
- Center for Neuroplasticity and Pain, Aalborg University, Aalborg, Denmark
| | - Rocco Giordano
- Center for Neuroplasticity and Pain, Aalborg University, Aalborg, Denmark
| | - Claus Vinter Bødker Hviid
- Department of Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Carsten Dahl Mørch
- Integrative Neuroscience, Aalborg University, Aalborg, Denmark
- Center for Neuroplasticity and Pain, Aalborg University, Aalborg, Denmark
| | - Pall Karlsson
- Danish Pain Research Center, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | | | - Lars Arendt-Nielsen
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
- Center for Neuroplasticity and Pain, Aalborg University, Aalborg, Denmark
- Mech-Sense, Department of Gastroenterology, Aalborg University Hospital, Aalborg, Denmark
| | - Niels Ejskjaer
- Steno Diabetes Center North Denmark, Aalborg University Hospital, Aalborg, Denmark
- Department Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Endocrinology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
15
|
Eldabe S, Nevitt S, Bentley A, Mekhail NA, Gilligan C, Billet B, Staats PS, Maden M, Soliday N, Leitner A, Duarte RV. Response to "Competing Narratives: Moving the Field Forward on Spinal Cord Stimulation". Clin J Pain 2024; 40:557-560. [PMID: 39023036 DOI: 10.1097/ajp.0000000000001232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024]
Affiliation(s)
- Sam Eldabe
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough
| | - Sarah Nevitt
- Centre for Reviews and Dissemination University of York, York
| | | | - Nagy A Mekhail
- Evidence-Based Pain Management Research, Cleveland Clinic, Cleveland, OH
| | | | | | | | - Michelle Maden
- Department of Health Data Science University of Liverpool, Liverpool, UK
| | - Nicole Soliday
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Angela Leitner
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Rui V Duarte
- Department of Health Data Science University of Liverpool, Liverpool, UK
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| |
Collapse
|
16
|
de Vos CC, Meier K. Spinal cord stimulation for the treatment of chronic pain. Nat Rev Neurol 2024; 20:447-448. [PMID: 38951599 DOI: 10.1038/s41582-024-00981-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Affiliation(s)
- Cecile C de Vos
- Center for Pain Medicine, Department of Anesthesiology, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Kaare Meier
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
- Department of Anesthesiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
17
|
Nurmikko T, Mugan D, Leitner A, Huygen FJPM. Quantitative Sensory Testing in Spinal Cord Stimulation: A Narrative Review. Neuromodulation 2024; 27:1026-1034. [PMID: 38639705 DOI: 10.1016/j.neurom.2024.03.005] [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: 01/18/2024] [Revised: 03/20/2024] [Accepted: 03/24/2024] [Indexed: 04/20/2024]
Abstract
OBJECTIVES Quantitative sensory testing (QST) has been used for decades to study sensory abnormalities in multiple conditions in which the somatosensory system is compromised, including pain. It is commonly used in pharmacologic studies on chronic pain but less so in conjunction with neuromodulation. This review aims to assess the utility of QST in spinal cord stimulation (SCS) protocols. MATERIALS AND METHODS For this narrative review, we searched PubMed for records of studies in which sensory testing has been performed as part of a clinical study on SCS from 1975 onward until October 2023. We focused on studies in which QST has been used to explore the effect of SCS on neuropathic, neuropathic-like, or mixed pain. RESULTS Our search identified 22 useful studies, all small and exploratory, using heterogeneous methods. Four studies used the full battery of validated German Research Network on Neuropathic Pain QST. There is emerging evidence that assessment dynamic mechanical allodynia (eight studies), and mechanical/thermal temporal summation of pain (eight studies) may have a role in quantifying the response to various SCS waveforms. There also were sporadic reports of improvement of sensory deficits in a proportion of patients with neuropathic pain that warrant further study. CONCLUSIONS We recommend the adoption of QST into future clinical research protocols, using either the full QST protocol or a less time-demanding short-form QST.
Collapse
Affiliation(s)
- Turo Nurmikko
- Department of Pain Medicine, The Walton Centre NHS Trust, Liverpool, UK.
| | - Dave Mugan
- Saluda Medical Europe Ltd, Harrogate, UK
| | - Angela Leitner
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Frank J P M Huygen
- Center for Pain Medicine, Erasmus University Medical Center, Rotterdam and UMCU, Utrecht, The Netherlands
| |
Collapse
|
18
|
Farnes N, Granan LP, Jacobsen HB, Stubhaug A, Vambheim SM. Associations of long-term clinical recovery and improved quality of life across ICD-11 chronic pain categories in a real-world registry study. Eur J Pain 2024; 28:1171-1184. [PMID: 38372480 DOI: 10.1002/ejp.2250] [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: 09/30/2023] [Revised: 01/15/2024] [Accepted: 02/02/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND There is little knowledge of what factors are needed for successful chronic pain management. We aim to identify psychosocial and treatment predictors of clinical recovery and improved quality of life (QOL) at 12-month follow-up across three chronic pain groups, based on the International Classification of Diseases-11: neuropathic pain, secondary non-neuropathic pain, and primary pain. Furthermore, we investigate baseline differences across diagnostic groups. METHODS The sample included baseline and 12-month follow-up data from 1056 chronic pain patients from the Oslo University Hospital's Pain Registry. Logistic regression models investigated longitudinal associations between psychosocial and treatment characteristics, and the outcome measures clinical recovery and improved QOL. Characteristics were compared across the diagnostic groups. RESULTS Increased odds of clinical recovery and improved QOL were seen in patients receiving invasive treatment (OR = 8.04, 95% CI = 3.50-19.40; OR = 5.47, 95% CI = 2.42-12.86), while decreased odds of clinical recovery were seen for secondary non-neuropathic pain patients with pain-related disability (0.05, 95% CI = 0.01-0.29). In comparing baseline characteristics, neuropathic pain patients had lower QOL, and more severe insomnia compared to the other groups. CONCLUSION Invasive treatment modalities were strongly associated with clinical recovery and improved QOL. Although this could be due to patient selection, it does warrant further examination as an intervention alternative for chronic pain. Intervention efficacy, risk factors and predictors of clinical recovery across diagnostic groups should be further investigated through longitudinal RCTs. SIGNIFICANCE This observational study indicates a potential advantage in sustained recovery for pre-selected individuals with chronic pain who undergo invasive treatments. The relationship between sustained recovery and psychosocial factors differs across neuropathic, secondary non-neuropathic, and primary pain patients. This suggests that employing ICD-11 for classifying patients into mechanistically distinct pain groups could inform the evaluation and management of chronic pain. Furthermore, factors previously identified as negative indicators for long-term outcomes in chronic pain cohorts were not clinically significant in this study.
Collapse
Affiliation(s)
- Nadine Farnes
- Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lars-Petter Granan
- Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Henrik B Jacobsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Audun Stubhaug
- Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Sara M Vambheim
- Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
19
|
Meier K, de Vos CC, Bordeleau M, van der Tuin S, Billet B, Ruland T, Blichfeldt-Eckhardt MR, Winkelmüller M, Gulisano HA, Gatzinsky K, Knudsen AL, Hedemann Sørensen JC, Milidou I, Cottin SC. Examining the Duration of Carryover Effect in Patients With Chronic Pain Treated With Spinal Cord Stimulation (EChO Study): An Open, Interventional, Investigator-Initiated, International Multicenter Study. Neuromodulation 2024; 27:887-898. [PMID: 38456888 DOI: 10.1016/j.neurom.2024.01.002] [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: 10/27/2023] [Revised: 12/17/2023] [Accepted: 01/16/2024] [Indexed: 03/09/2024]
Abstract
OBJECTIVES Spinal cord stimulation (SCS) is a surgical treatment for severe, chronic, neuropathic pain. It is based on one to two lead(s) implanted in the epidural space, stimulating the dorsal column. It has long been assumed that when deactivating SCS, there is a variable interval before the patient perceives the return of the pain, a phenomenon often termed echo or carryover effect. Although the carryover effect has been problematized as a source of error in crossover studies, no experimental investigation of the effect has been published. This open, prospective, international multicenter study aimed to systematically document, quantify, and investigate the carryover effect in SCS. MATERIALS AND METHODS Eligible patients with a beneficial effect from their SCS treatment were instructed to deactivate their SCS device in a home setting and to reactivate it when their pain returned. The primary outcome was duration of carryover time defined as the time interval from deactivation to reactivation. Central clinical parameters (age, sex, indication for SCS, SCS treatment details, pain score) were registered and correlated with carryover time using nonparametric tests (Mann-Whitney/Kruskal-Wallis) for categorical data and linear regression for continuous data. RESULTS In total, 158 patients were included in the analyses. A median carryover time of five hours was found (interquartile range 2.5;21 hours). Back pain as primary indication for SCS, high-frequency stimulation, and higher pain score at the time of deactivation were correlated with longer carryover time. CONCLUSIONS This study confirms the existence of the carryover effect and indicates a remarkably high degree of interindividual variation. The results suggest that the magnitude of carryover may be correlated to the nature of the pain condition and possibly stimulation paradigms. CLINICAL TRIAL REGISTRATION The Clinicaltrials.gov registration number for the study is NCT03386058.
Collapse
Affiliation(s)
- Kaare Meier
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark; Department of Anesthesiology, Aarhus University Hospital, Aarhus, Denmark; Center for Experimental Neuroscience (CENSE), Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Cecile C de Vos
- Center for Pain Medicine, Department of Anesthesiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Martine Bordeleau
- Research Centre on Aging, CIUSSS de l'Estrie-CHUS, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Sharon van der Tuin
- Department of Neurosurgery, Medisch Spectrum Twente, Enschede, The Netherlands
| | - Bart Billet
- Department of Anesthesiology, AZ Delta, Roeselare, Belgium
| | | | | | | | | | - Kliment Gatzinsky
- Department of Neurosurgery, Sahlgrenska University Hospital, Göteborg, Sweden
| | - Anne Lene Knudsen
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Christian Hedemann Sørensen
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark; Center for Experimental Neuroscience (CENSE), Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Ioanna Milidou
- Department of Pediatrics and Adolescent Medicine, Regional Hospital West Jutland, Herning, Denmark; Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
20
|
Wondwossen Y, Patzkowski MS, Amoako MY, Lawson BK, Velosky AG, Soto AT, Highland KB. Spinal Cord Stimulator Inequities Within the US Military Health System. Neuromodulation 2024; 27:916-922. [PMID: 38971583 DOI: 10.1016/j.neurom.2023.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/20/2023] [Accepted: 03/13/2023] [Indexed: 07/08/2024]
Abstract
OBJECTIVES Although studies have described inequities in spinal cord stimulation (SCS) receipt, there is a lack of information to inform system-level changes to support health care equity. This study evaluated whether Black patients exhaust more treatment options than do White patients, before receiving SCS. MATERIALS AND METHODS This retrospective cohort study included claims data of Black and non-Latinx White patients who were active-duty service members or military retirees who received a persistent spinal pain syndrome (PSPS) diagnosis associated with back surgery within the US Military Health System, January 2017 to January 2020 (N = 8753). A generalized linear model examined predictors of SCS receipt within two years of diagnosis, including the interaction between race and number of pain-treatment types received. RESULTS In the generalized linear model, Black patients (10.3% [8.7%, 12.0%]) were less likely to receive SCS than were White patients (13.6% [12.7%, 14.6%]) The interaction term was significant; White patients who received zero to three different types of treatments were more likely to receive SCS than were Black patients who received zero to three treatments, whereas Black and White patients who received >three treatments had similar likelihoods of receiving a SCS. CONCLUSIONS In a health care system with intended universal access, White patients diagnosed with PSPS tried fewer treatment types before receiving SCS, whereas the number of treatment types tried was not significantly related to SCS receipt in Black patients. Overall, Black patients received SCS less often than did White patients. Findings indicate the need for structured referral pathways, provider evaluation on equity metrics, and top-down support.
Collapse
Affiliation(s)
- Ysehak Wondwossen
- School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Michael S Patzkowski
- Department of Anesthesiology, Brooke Army Medical Center, Fort Sam Houston, TX, USA; Department of Anesthesiology, Uniformed Services University, Bethesda, MD, USA
| | - Maxwell Y Amoako
- Enterprise Intelligence and Data Solutions program office, Program Executive Office, Defense Healthcare Management Systems, San Antonio, TX, USA; Defense and Veterans Center for Integrative Pain Management, Department of Anesthesiology, Uniformed Services University, Bethesda, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Bryan K Lawson
- Department of Orthopedic Surgery, Brooke Army Medical Center, Fort Sam Houston, TX, USA
| | - Alexander G Velosky
- Enterprise Intelligence and Data Solutions program office, Program Executive Office, Defense Healthcare Management Systems, San Antonio, TX, USA; Defense and Veterans Center for Integrative Pain Management, Department of Anesthesiology, Uniformed Services University, Bethesda, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Adam T Soto
- Department of Anesthesiology, Uniformed Services University, Bethesda, MD, USA; Department of Anesthesiology, Tripler Army Medical Center, Honolulu, HI, USA
| | - Krista B Highland
- Department of Anesthesiology, Uniformed Services University, Bethesda, MD, USA.
| |
Collapse
|
21
|
Buonanno P, Servillo G, Visser-Vandewalle V, Matis G. 10 kHz stimulation as rescue therapy for spinal cord stimulation trial failure or loss of efficacy: A retrospective study. Pain Pract 2024; 24:845-851. [PMID: 38558532 DOI: 10.1111/papr.13369] [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: 12/18/2023] [Revised: 02/25/2024] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Spinal cord stimulation (SCS) is currently used for the management of pain of different origin, and since its inception, many waveforms have been developed. Some patients experience no pain relief already during SCS trial, while other patients go through a loss of efficacy due to habituation after a variable period of satisfying pain control. Our retrospective study represents the first report exploring the potential role of 10 kHz stimulation as rescue therapy for patients who did not benefit not only from conventional stimulation but even from other waveforms during SCS trial or follow-up. METHODS This study was conducted in Germany; we retrospectively enrolled patients with no pain relief during SCS trial or with loss of efficacy of other waveforms over time; and we recorded visual analogic scale (VAS), Oswestry Disability Index (ODI), and daily opioid consumption expressed as morphine milligram equivalents (MME), right before and 12 months after the switching to 10 kHz simulation. RESULTS The rate of successful switching to 10 kHz stimulation was comparable in patients enrolled during the SCS trial and during the follow-up (43% vs. 40%, respectively); notably, the highest rate of failed rescue was recorded in case of persistent spinal pain syndrome (PSPS) II. Patients who responded to the switching showed a significant improvement in VAS and ODI after 12 months of treatment compared to baseline (3.6 ± 1.0 vs. 8.2 ± 0.9, p < 0.00001 and 34.0 ± 7.8 vs. 64.3 ± 8.7, p < 0.0001, respectively), whereas there was no reduction in the consumption of opioids in terms of MME (3 (0-16) vs. 5 (0-8.75), p = 0.1003). CONCLUSIONS Rescue therapy with 10 kHz stimulation could be an important strategy to avoid SCS explant in both patients non-responsive during trial or experiencing a loss of efficacy during the years with other waveforms.
Collapse
Affiliation(s)
- Pasquale Buonanno
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples "Federico II", Naples, Italy
| | - Giuseppe Servillo
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples "Federico II", Naples, Italy
| | - Veerle Visser-Vandewalle
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Georgios Matis
- Department of Stereotactic and Functional Neurosurgery, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
22
|
Klasova J, Hussain N, Umer I, Al-Hindawi A, ElSaban M, Lahori S, D'Souza RS. Emotional and psychosocial function after dorsal column spinal cord stimulator implantation: a systematic review and meta-analysis. Reg Anesth Pain Med 2024:rapm-2024-105523. [PMID: 38942426 DOI: 10.1136/rapm-2024-105523] [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: 03/30/2024] [Accepted: 06/08/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND The efficacy of spinal cord stimulation (SCS) in chronic pain studies is traditionally assessed by pain scores, which do not reflect the multidimensional nature of pain perception. Despite the evidence of SCS's influence on emotional functioning comprehensive assessments of its effect remain lacking. OBJECTIVE To assess changes in emotional and psychosocial functioning in patients who underwent SCS implantation for chronic pain. EVIDENCE REVIEW Ovid MEDLINE, EMBASE, PsychINFO, Cochrane CENTRAL and Scopus databases were searched for original peer-reviewed publications reporting emotional functioning after SCS. The primary outcomes were a pooled mean difference (MD) in anxiety, depression, global functioning, mental well-being and pain catastrophizing at 12 months. The Grading of Recommendation, Assessment, Development, and Evaluation (GRADE) was used to determine the quality of evidence. FINDINGS Thirty-two studies were included in the primary analysis. Statistically significant improvements were observed in anxiety (MD -2.16; 95% CI -2.84 to -1.49; p<0.001), depression (MD -4.66; 95% CI -6.26 to -3.06; p<0.001), global functioning (MD 20.30; 95% CI 14.69 to 25.90; p<0.001), mental well-being (MD 4.95; 95% CI 3.60 to 6.31; p<0.001), and pain catastrophizing (MD -12.09; 95% CI -14.94 to -9.23; p<0.001). Subgroup analyses revealed differences in Global Assessment of Functioning and mental well-being based on study design and in depression based on waveform paradigm. CONCLUSION The results highlight the statistically and clinically significant improvements in emotional and psychosocial outcomes in patients with chronic pain undergoing SCS therapy. However, these results need to be interpreted with caution due to the very low certainty of evidence per the GRADE criteria. PROSPERO REGISTRATION CRD42023446326.
Collapse
Affiliation(s)
- Johana Klasova
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Nasir Hussain
- Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Ibrahim Umer
- Department of Anesthesiology, St Joseph's University Medical Center, Paterson, New Jersey, USA
| | - Ahmed Al-Hindawi
- Royal College of Surgeons in Ireland Medical University of Bahrain, Al Muharraq, Bahrain
| | - Mariam ElSaban
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Simmy Lahori
- Department of Hematology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryan S D'Souza
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
23
|
Sokal P, Palus D, Jabłońska M, Puk O, Kieronska-Siwak S. Spinal Cord Stimulation for Central Neuropathic Pain After Spinal Cord Injury: A Single-Center Case Series. J Pain Res 2024; 17:2029-2035. [PMID: 38881761 PMCID: PMC11177860 DOI: 10.2147/jpr.s462587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Purpose Central neuropathic pain (CNP) following spinal cord injury (SCI) presents a formidable therapeutic challenge, affecting over 50% of the patients post-SCI. For those who experience CNP, conventional treatments often prove insufficient. Spinal cord stimulation (SCS) emerges as a potential intervention for chronic pain after SCI that is unresponsive to pharmacotherapy and supportive measures. However, the efficacy of SCS in alleviating CNP is notably limited. The objective of our study was to evaluate novel stimulation paradigms in SCS for patients with severe CNP after SCI, based on our extensive experience. Patients and Methods From a pool of 112 patients treated with SCS for chronic neuropathic pain in the Department of Neurosurgery and Neurology, we selected eight individuals (4 males and 4 females) with CNP for our case series. Burst and high frequency SCS was applied. The assessment involved utilizing the Numeric Rating Scale (NRS), the Neuropathic Pain Symptom Inventory (NPSI), and the EQ-5D quality of life scale before surgery and during a 12-month follow-up period. Results Over the course of the one-year follow-up, only two patients experienced satisfactory relief from pain, demonstrating the effectiveness of the stimulation. Moreover, high-frequency and burst SCS failed to show improvement in the remaining six patients. Conclusion Our findings suggest that, despite the incorporation of new stimulation paradigms such as burst stimulation and high-frequency stimulation, SCS does not exhibit significant effectiveness in treating neuropathic pain in patients after SCI. These findings highlight the ongoing challenge of treating CNP and emphasize the importance of investigating alternative therapeutic strategies for this group.
Collapse
Affiliation(s)
- Paweł Sokal
- Department of Neurosurgery and Neurology, Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, 85-168, Poland
| | - Damian Palus
- Department of Neurosurgery and Neurology, Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, 85-168, Poland
| | - Magdalena Jabłońska
- Doctoral School Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, 85-168, Poland
| | - Oskar Puk
- Doctoral School Collegium Medicum Nicolaus Copernicus University, Bydgoszcz, 85-168, Poland
| | - Sara Kieronska-Siwak
- Department of Neurosurgery and Neurology, Faculty of Health Sciences, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, 85-168, Poland
| |
Collapse
|
24
|
Cao B, Xu Q, Shi Y, Zhao R, Li H, Zheng J, Liu F, Wan Y, Wei B. Pathology of pain and its implications for therapeutic interventions. Signal Transduct Target Ther 2024; 9:155. [PMID: 38851750 PMCID: PMC11162504 DOI: 10.1038/s41392-024-01845-w] [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: 05/12/2023] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 06/10/2024] Open
Abstract
Pain is estimated to affect more than 20% of the global population, imposing incalculable health and economic burdens. Effective pain management is crucial for individuals suffering from pain. However, the current methods for pain assessment and treatment fall short of clinical needs. Benefiting from advances in neuroscience and biotechnology, the neuronal circuits and molecular mechanisms critically involved in pain modulation have been elucidated. These research achievements have incited progress in identifying new diagnostic and therapeutic targets. In this review, we first introduce fundamental knowledge about pain, setting the stage for the subsequent contents. The review next delves into the molecular mechanisms underlying pain disorders, including gene mutation, epigenetic modification, posttranslational modification, inflammasome, signaling pathways and microbiota. To better present a comprehensive view of pain research, two prominent issues, sexual dimorphism and pain comorbidities, are discussed in detail based on current findings. The status quo of pain evaluation and manipulation is summarized. A series of improved and innovative pain management strategies, such as gene therapy, monoclonal antibody, brain-computer interface and microbial intervention, are making strides towards clinical application. We highlight existing limitations and future directions for enhancing the quality of preclinical and clinical research. Efforts to decipher the complexities of pain pathology will be instrumental in translating scientific discoveries into clinical practice, thereby improving pain management from bench to bedside.
Collapse
Affiliation(s)
- Bo Cao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qixuan Xu
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Yajiao Shi
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China
| | - Ruiyang Zhao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Hanghang Li
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Jie Zheng
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China
| | - Fengyu Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China.
| | - You Wan
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission, Peking University, Beijing, 100191, China.
| | - Bo Wei
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
25
|
Mekhail NA, Levy RM, Deer TR, Kapural L, Li S, Amirdelfan K, Pope JE, Hunter CW, Rosen SM, Costandi SJ, Falowski SM, Burgher AH, Gilmore CA, Qureshi FA, Staats PS, Scowcroft J, McJunkin T, Carlson J, Kim CK, Yang MI, Stauss T, Petersen EA, Hagedorn JM, Rauck R, Kallewaard JW, Baranidharan G, Taylor RS, Poree L, Brounstein D, Duarte RV, Gmel GE, Gorman R, Gould I, Hanson E, Karantonis DM, Khurram A, Leitner A, Mugan D, Obradovic M, Ouyang Z, Parker J, Single P, Soliday N. ECAP-controlled closed-loop versus open-loop SCS for the treatment of chronic pain: 36-month results of the EVOKE blinded randomized clinical trial. Reg Anesth Pain Med 2024; 49:346-354. [PMID: 37640452 PMCID: PMC11103285 DOI: 10.1136/rapm-2023-104751] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/13/2023] [Indexed: 08/31/2023]
Abstract
INTRODUCTION The evidence for spinal cord stimulation (SCS) has been criticized for the absence of blinded, parallel randomized controlled trials (RCTs) and limited evaluations of the long-term effects of SCS in RCTs. The aim of this study was to determine whether evoked compound action potential (ECAP)-controlled, closed-loop SCS (CL-SCS) is associated with better outcomes when compared with fixed-output, open-loop SCS (OL-SCS) 36 months following implant. METHODS The EVOKE study was a multicenter, participant-blinded, investigator-blinded, and outcome assessor-blinded, randomized, controlled, parallel-arm clinical trial that compared ECAP-controlled CL-SCS with fixed-output OL-SCS. Participants with chronic, intractable back and leg pain refractory to conservative therapy were enrolled between January 2017 and February 2018, with follow-up through 36 months. The primary outcome was a reduction of at least 50% in overall back and leg pain. Holistic treatment response, a composite outcome including pain intensity, physical and emotional functioning, sleep, and health-related quality of life, and objective neural activation was also assessed. RESULTS At 36 months, more CL-SCS than OL-SCS participants reported ≥50% reduction (CL-SCS=77.6%, OL-SCS=49.3%; difference: 28.4%, 95% CI 12.8% to 43.9%, p<0.001) and ≥80% reduction (CL-SCS=49.3%, OL-SCS=31.3%; difference: 17.9, 95% CI 1.6% to 34.2%, p=0.032) in overall back and leg pain intensity. Clinically meaningful improvements from baseline were observed at 36 months in both CL-SCS and OL-SCS groups in all other patient-reported outcomes with greater levels of improvement with CL-SCS. A greater proportion of patients with CL-SCS were holistic treatment responders at 36-month follow-up (44.8% vs 28.4%), with a greater cumulative responder score for CL-SCS patients. Greater neural activation and accuracy were observed with CL-SCS. There were no differences between CL-SCS and OL-SCS groups in adverse events. No explants due to loss of efficacy were observed in the CL-SCS group. CONCLUSION This long-term evaluation with objective measurement of SCS therapy demonstrated that ECAP-controlled CL-SCS resulted in sustained, durable pain relief and superior holistic treatment response through 36 months. Greater neural activation and increased accuracy of therapy delivery were observed with ECAP-controlled CL-SCS than OL-SCS. TRIAL REGISTRATION NUMBER NCT02924129.
Collapse
Affiliation(s)
- Nagy A Mekhail
- Department of Pain Management, Cleveland Clinic, Cleveland, Ohio, USA
| | - Robert M Levy
- Neurosurgical Services, Anesthesia Pain Care Consultants, Boca Raton, Florida, USA
| | - Timothy R Deer
- Spine and Nerve Center of the Virginias, West Virginia University - Health Sciences Campus, Morgantown, West Virginia, USA
| | - Leonardo Kapural
- Carolinas Pain Institute, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Sean Li
- Premier Pain Centers, Shrewsbury, New Jersey, USA
| | - Kasra Amirdelfan
- Research, Integrated Pain Management Medical Group Inc, Walnut Creek, California, USA
| | - Jason E Pope
- Evolve Restorative Center, Santa Rosa, California, USA
| | - Corey W Hunter
- Ainsworth Institute of Pain Management, New York, New York, USA
| | - Steven M Rosen
- Delaware Valley Pain and Spine Institute, Trevose, Pennsylvania, USA
| | - Shrif J Costandi
- Department of Pain Management, Cleveland Clinic, Cleveland, Ohio, USA
| | - Steven M Falowski
- Argires-Marotti Neurosurgical Associates of Lancaster, Lancaster, Pennsylvania, USA
| | | | - Christopher A Gilmore
- Center for Clinical Research, Carolinas Pain Institute, Winston-Salem, North Carolina, USA
| | | | | | | | | | | | - Christopher K Kim
- Spine and Nerve Center of the Virginias, West Virginia University - Health Sciences Campus, Morgantown, West Virginia, USA
| | | | - Thomas Stauss
- Pain Physicians of Wisconsin, Milwaukee, Wisconsin, USA
| | - Erika A Petersen
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | - Richard Rauck
- Center for Clinical Research, Carolinas Pain Institute, Winston-Salem, North Carolina, USA
| | - Jan W Kallewaard
- Anesthesiology and Pain Medicine, Rijnstate Hospital, Arnhem, The Netherlands
- Anesthesiology and Pain Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | | | - Rod S Taylor
- Institute of Health and Well Being, University of Glasgow, Glasgow, UK
| | - Lawrence Poree
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, California, USA
| | - Dan Brounstein
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Rui V Duarte
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
- Health Data Science, University of Liverpool, Liverpool, UK
| | - Gerrit E Gmel
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Robert Gorman
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Ian Gould
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Erin Hanson
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | | | - Abeer Khurram
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Angela Leitner
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Dave Mugan
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Milan Obradovic
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Zhonghua Ouyang
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - John Parker
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Peter Single
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Nicole Soliday
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| |
Collapse
|
26
|
Eldabe S, Nevitt S, Copley S, Maden M, Goudman L, Hayek S, Mekhail N, Moens M, Rigoard P, Duarte R. Does industry funding and study location impact findings from randomized controlled trials of spinal cord stimulation? A systematic review and meta-analysis. Reg Anesth Pain Med 2024; 49:272-284. [PMID: 37611944 DOI: 10.1136/rapm-2023-104674] [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: 05/10/2023] [Accepted: 08/13/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND/IMPORTANCE Concerns have been raised that effects observed in studies of spinal cord stimulation (SCS) funded by industry have not been replicated in non-industry-funded studies and that findings may differ based on geographical location where the study was conducted. OBJECTIVE To investigate the impact of industry funding and geographical location on pain intensity, function, health-related quality of life and adverse events reported in randomized controlled trials (RCTs) of SCS. EVIDENCE REVIEW Systematic review conducted using MEDLINE, CENTRAL, EMBASE and WikiStim databases until September 2022. Parallel-group RCTs evaluating SCS for patients with neuropathic pain were included. Results of studies were combined in random-effects meta-analysis using the generic-inverse variance method. Subgroup meta-analyses were conducted according to funding source and study location. Risk of bias was assessed using Cochrane RoB 2.0 tool. FINDINGS Twenty-nine reports of 17 RCTs (1823 participants) were included. For the comparison of SCS with usual care, test for subgroup differences indicate no significant differences (p=0.48, moderate certainty evidence) in pain intensity score at 6 months for studies with no funding or funding not disclosed (pooled mean difference (MD) -1.96 (95% CI -3.23 to -0.69; 95% prediction interval (PI) not estimable, I2=0%, τ2=0)), industry funding (pooled MD -2.70 (95% CI -4.29 to -1.11; 95% PI -8.75 to 3.35, I2=97%, τ2=2.96) or non-industry funding (MD -3.09 (95% CI -4.47 to -1.72); 95% PI, I2 and τ2 not applicable). Studies with industry funding for the comparison of high-frequency SCS (HF-SCS) with low-frequency SCS (LF-SCS) showed statistically significant advantages for HF-SCS compared to LF-SCS while studies with no funding showed no differences between HF-SCS and LF-SCS (low certainty evidence). CONCLUSION All outcomes of SCS versus usual care were not significantly different between studies funded by industry and those independent from industry. Pain intensity score and change in pain intensity from baseline for comparisons of HF-SCS to LF-SCS seem to be impacted by industry funding.
Collapse
Affiliation(s)
- Sam Eldabe
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Sarah Nevitt
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Sue Copley
- Anaesthesia and Pain Management, James Cook University Hospital, Middlesbrough, UK
| | - Michelle Maden
- Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Lisa Goudman
- Department of Neurosurgery, UZ Brussel, Brussel, Belgium
| | - Salim Hayek
- Anesthesiology, Case Western Reserve University, Unviersity Hospitals, Cleveland, Ohio, USA
| | | | - Maarten Moens
- Department of Neurosurgery, UZ Brussel, Brussel, Belgium
| | - Phillipe Rigoard
- PRISMATICS Lab, Poitiers, France
- Department of Neurosurgery, Poitiers University Hospital, Poitiers, France
| | | |
Collapse
|
27
|
Tieppo Francio V, Alm J, Leavitt L, Mok D, Yoon BV, Nazir N, Lam C, Latif U, Sowder T, Braun E, Sack A, Khan T, Sayed D. Variables associated with nonresponders to high-frequency (10 kHz) spinal cord stimulation. Pain Pract 2024; 24:584-599. [PMID: 38078593 DOI: 10.1111/papr.13328] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
INTRODUCTION The use of spinal cord stimulation (SCS) therapy to treat chronic pain continues to rise. Optimal patient selection remains one of the most important factors for SCS success. However, despite increased utilization and the existence of general indications, predicting which patients will benefit from neuromodulation remains one of the main challenges for this therapy. Therefore, this study aims to identify the variables that may correlate with nonresponders to high-frequency (10 kHz) SCS to distinguish the subset of patients less likely to benefit from this intervention. MATERIALS AND METHODS This was a retrospective single-center observational study of patients who underwent 10 kHz SCS implant. Patients were divided into nonresponders and responders groups. Demographic data and clinical outcomes were collected at baseline and statistical analysis was performed for all continuous and categorical variables between the two groups to calculate statistically significant differences. RESULTS The study population comprised of 237 patients, of which 67.51% were responders and 32.49% were nonresponders. There was a statistically significant difference of high levels of kinesiophobia, high self-perceived disability, greater pain intensity, and clinically relevant pain catastrophizing at baseline in the nonresponders compared to the responders. A few variables deemed potentially relevant, such as age, gender, history of spinal surgery, diabetes, alcohol use, tobacco use, psychiatric illness, and opioid utilization at baseline were not statistically significant. CONCLUSION Our study is the first in the neuromodulation literature to raise awareness to the association of high levels of kinesiophobia preoperatively in nonresponders to 10 kHz SCS therapy. We also found statistically significant differences with greater pain intensity, higher self-perceived disability, and clinically relevant pain catastrophizing at baseline in the nonresponders relative to responders. It may be appropriate to screen for these factors preoperatively to identify patients who are less likely to respond to SCS. If these modifiable risk factors are present, it might be prudent to consider a pre-rehabilitation program with pain neuroscience education to address these factors prior to SCS therapy, to enhance successful outcomes in neuromodulation.
Collapse
Affiliation(s)
- Vinicius Tieppo Francio
- Department of Physical Medicine and Rehabilitation, The University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - John Alm
- Department of Physical Medicine and Rehabilitation, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Logan Leavitt
- Department of Physical Medicine and Rehabilitation, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Daniel Mok
- Department of Physical Medicine and Rehabilitation, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - B Victor Yoon
- Department of Physical Medicine and Rehabilitation, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Niaman Nazir
- Department of Population Health, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Christopher Lam
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Usman Latif
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Timothy Sowder
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Edward Braun
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Andrew Sack
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Talal Khan
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Dawood Sayed
- Department of Anesthesiology and Pain Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
28
|
Mekhail NA, Levy RM, Deer TR, Kapural L, Li S, Amirdelfan K, Hunter CW, Rosen SM, Costandi SJ, Falowski SM, Burgher AH, Pope JE, Gilmore CA, Qureshi FA, Staats PS, Scowcroft J, McJunkin T, Kim CK, Yang MI, Stauss T, Rauck R, Duarte RV, Soliday N, Leitner A, Hanson E, Ouyang Z, Mugan D, Poree L. Neurophysiological outcomes that sustained clinically significant improvements over 3 years of physiologic ECAP-controlled closed-loop spinal cord stimulation for the treatment of chronic pain. Reg Anesth Pain Med 2024:rapm-2024-105370. [PMID: 38490687 DOI: 10.1136/rapm-2024-105370] [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: 02/02/2024] [Accepted: 02/20/2024] [Indexed: 03/17/2024]
Abstract
INTRODUCTION A novel, spinal cord stimulation (SCS) system with a physiologic closed-loop (CL) feedback mechanism controlled by evoked compound action potentials (ECAPs) enables the optimization of physiologic neural dose and the accuracy of the stimulation, not possible with any other commercially available SCS systems. The report of objective spinal cord measurements is essential to increase the transparency and reproducibility of SCS therapy. Here, we report a cohort of the EVOKE double-blind randomized controlled trial treated with CL-SCS for 36 months to evaluate the ECAP dose and accuracy that sustained the durability of clinical improvements. METHODS 41 patients randomized to CL-SCS remained in their treatment allocation and were followed up through 36 months. Objective neurophysiological data, including measures of spinal cord activation, were analyzed. Pain relief was assessed by determining the proportion of patients with ≥50% and ≥80% reduction in overall back and leg pain. RESULTS The performance of the feedback loop resulted in high-dose accuracy by keeping the elicited ECAP within 4µV of the target ECAP set on the system across all timepoints. Percent time stimulating above the ECAP threshold was >98%, and the ECAP dose was ≥19.3µV. Most patients obtained ≥50% reduction (83%) and ≥80% reduction (59%) in overall back and leg pain with a sustained response observed in the rates between 3-month and 36-month follow-up (p=0.083 and p=0.405, respectively). CONCLUSION The results suggest that a physiological adherence to supra-ECAP threshold therapy that generates pain inhibition provided by ECAP-controlled CL-SCS leads to durable improvements in pain intensity with no evidence of loss of therapeutic effect through 36-month follow-up.
Collapse
Affiliation(s)
- Nagy A Mekhail
- Evidence-Based Pain Management Research, Cleveland Clinic, Cleveland, Ohio, USA
| | - Robert M Levy
- Anesthesia Pain Care Consultants, Boca Raton, Florida, USA
| | - Timothy R Deer
- Spine and Nerve Center of the Virginias, Charleston, West Virginia, USA
| | | | - Sean Li
- National Spine and Pain Centers, Shrewsbury, New Jersey, USA
| | - Kasra Amirdelfan
- Integrated Pain Management Medical Group Inc, Walnut Creek, California, USA
| | - Corey W Hunter
- Ainsworth Institute of Pain Management, New York, New York, USA
| | - Steven M Rosen
- Delaware Valley Pain and Spine Institute, Trevose, Pennsylvania, USA
| | - Shrif J Costandi
- Evidence-Based Pain Management Research, Cleveland Clinic, Cleveland, Ohio, USA
| | - Steven M Falowski
- Argires-Marotti Neurosurgical Associates of Lancaster, Lancaster, Pennsylvania, USA
| | | | - Jason E Pope
- Evolve Restorative Center, Santa Rosa, California, USA
| | | | | | - Peter S Staats
- National Spine and Pain Centers, Shrewsbury, New Jersey, USA
| | | | | | - Christopher K Kim
- Spine and Nerve Center of the Virginias, Charleston, West Virginia, USA
| | | | - Thomas Stauss
- Pain Physicians of Wisconsin, Milwaukee, Wisconsin, USA
| | - Richard Rauck
- Carolinas Pain Institute, Winston-Salem, North Carolina, USA
| | - Rui V Duarte
- Department of Health Data Science, University of Liverpool, Liverpool, UK
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Nicole Soliday
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Angela Leitner
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Erin Hanson
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Zhonghua Ouyang
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Dave Mugan
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Lawrence Poree
- University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
29
|
Kaijankoski H, Nissen M, Ikäheimo TM, von und zu Fraunberg M, Airaksinen O, Huttunen J. Neuropathic Pain Medication and Antidepressant Use after Disability Pension in Patients with Spinal Cord Stimulation for Persistent Spinal Pain Syndrome. Pain Res Manag 2024; 2024:4953758. [PMID: 38327724 PMCID: PMC10849812 DOI: 10.1155/2024/4953758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 02/09/2024]
Abstract
Background Treatment of persistent spinal pain syndrome (PSPS) is challenging. Chronic pain associated with PSPS can lead to an impaired ability to work. Objective To obtain information on whether receiving a disability pension (DP) affects pain and pain treatments in retiring working-age PSPS patients. Neuropathic pain medication and antidepressant use were considered as an indicator of neuropathic pain. Methods The study group comprised 129 consecutive PSPS patients with spinal cord stimulation (SCS) devices implanted at Kuopio University Hospital Neurosurgery between January 1, 1996, and December 31, 2014. Purchase data of gabapentinoids, tricyclic antidepressants, and serotonin-norepinephrine reuptake inhibitors from January 1995 to March 2016, as well as the data on working ability, were retrieved from national registries. Results The data showed that 28 of 129 (21.7%) SCS permanent patients had a DP, and 27 had a sufficient follow-up time (two years before and one year after DP). Most patients (61%) used neuropathic pain medications during the follow-up, while 44% used antidepressants. Most patients (70%, n = 19) retired because of dorsopathies. The dose of gabapentinoids started to increase before the DP; after the DP, the doses started to increase again after the decrease but remained at a lower level. Conclusions Neuropathic pain medication and antidepressant use suggest that pain continues after the DP-that is, pensioners continue to experience inconvenient chronic pain. Resources for patient care are therefore needed after the DP. However, the DP reduces the dose increase of gabapentinoids; the dose is higher immediately before retirement than at the end of the follow-up.
Collapse
Affiliation(s)
- Hanna Kaijankoski
- Departments of Physical and Rehabilitation Medicine, Kuopio University Hospital, PB 100, KYS 70029, Kuopio, Finland
- University of Eastern Finland, Kuopio, Finland
| | - Mette Nissen
- Neurosurgery, Kuopio University Hospital, PB 100, 70029 KYS, Kuopio, Finland
| | - Tiina-Mari Ikäheimo
- Neurosurgery, Kuopio University Hospital, PB 100, 70029 KYS, Kuopio, Finland
| | - Mikael von und zu Fraunberg
- Neurosurgery, Oulu University Hospital, Kajaanintie 50, Oulu 90220, Finland
- University of Oulu, Oulu, Finland
| | - Olavi Airaksinen
- Departments of Physical and Rehabilitation Medicine, Kuopio University Hospital, PB 100, KYS 70029, Kuopio, Finland
- University of Eastern Finland, Kuopio, Finland
| | - Jukka Huttunen
- Neurosurgery, Kuopio University Hospital, PB 100, 70029 KYS, Kuopio, Finland
| |
Collapse
|
30
|
Yeung AM, Huang J, Nguyen KT, Xu NY, Hughes LT, Agrawal BK, Ejskjaer N, Klonoff DC. Painful Diabetic Neuropathy: The Need for New Approaches. J Diabetes Sci Technol 2024; 18:159-167. [PMID: 36305521 PMCID: PMC10899841 DOI: 10.1177/19322968221132252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Painful diabetic neuropathy is a common vexing problem for people with diabetes and a costly problem for society. The pathophysiology is not well understood, and no safe and effective mechanistically-based treatment has been identified. Poor glycemic control is a risk factor for painful diabetic neuropathy. Excessive intraneuronal glucose in people with diabetes can be shunted away from physiological glycolysis into multiple pathological pathways associated with neuropathy and pain. The first three treatments that are traditionally offered consist of risk factor reduction, lifestyle modifications, and pharmacological therapy, which includes only three drugs that are approved for this indication by the United States Food and Drug Administration. All of these traditional treatments are often inadequate for relieving neuropathic pain, and thus, new approaches are needed. Modern devices based on neuromodulation technology, which act directly on the nervous system, have been recently cleared by the United States Food and Drug Administration for painful diabetic neuropathy and offer promise as next-in-line therapy when traditional therapies fail.
Collapse
Affiliation(s)
| | | | | | - Nicole Y. Xu
- Diabetes Technology Society, Burlingame, CA, USA
| | - Lorenzo T. Hughes
- Balance Health, San Francisco, CA, USA
- Mills-Peninsula Medical Center, Burlingame, CA, USA
| | | | - Niels Ejskjaer
- Steno Diabetes Center North Denmark and Department of Endocrinology, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - David C. Klonoff
- Diabetes Technology Society, Burlingame, CA, USA
- Diabetes Research Institute, Mills-Peninsula Medical Center, San Mateo, CA, USA
| |
Collapse
|
31
|
Rigoard P, Ounajim A, Moens M, Goudman L, Roulaud M, Lorgeoux B, Baron S, Nivole K, Many M, Lampert L, David R, Billot M. Should we Oppose or Combine Waveforms for Spinal Cord Stimulation in PSPS-T2 Patients? A Prospective Randomized Crossover Trial (MULTIWAVE Study). THE JOURNAL OF PAIN 2023; 24:2319-2339. [PMID: 37473903 DOI: 10.1016/j.jpain.2023.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/16/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023]
Abstract
Refractory persistent spinal pain syndrome after surgery (PSPS-T2) can be successfully addressed by spinal cord stimulation (SCS). While conventional stimulation generates paresthesia, recent systems enable the delivery of paresthesia-free stimulation. Studies have claimed non-inferiority/superiority of selected paresthesia-free stimulation compared with paresthesia-based stimulation, but the comparative efficacy between different waveforms still needs to be determined in a given patient. We designed a randomized controlled 3-month crossover trial to compare pain relief of paresthesia-based stimulation versus high frequency versus burst in 28 PSPS-T2 patients implanted with multiwave SCS systems. Our secondary objectives were to determine the efficacy of these 3 waveforms on pain surface, quality of life, functional capacity, psychological distress, and validated composite multidimensional clinical response index to provide holistic comparisons at 3-, 6-, 9-, and 15-month post-randomization. The preferred stimulation modality was documented during the follow-up periods. No difference between the waveforms was observed in this study (P = .08). SCS led to significant pain relief, quality of life improvement, improvement of multidimensional clinical response index, and of all other clinical outcomes at all follow-up visits. Forty-four percent of the patients chose to keep the paresthesia-based stimulation modality after the 15-month follow-up period. By giving the possibility to switch and/or to combine several waveforms, the overall rate of SCS responders further increased with 25%. In this study, high frequency or burst do not appear superior to paresthesia-based stimulation, wherefore paresthesia-based stimulation should still be considered as a valid option. However, combining paresthesia-based stimulation with paresthesia-free stimulation, through personalized multiwave therapy, might significantly improve SCS responses. PERSPECTIVE: This article assesses clinical SCS efficacy on pain relief, by comparing paresthesia-based stimulation and paresthesia-free stimulation (including high frequency and burst) modalities in patient presenting with PSPS-T2. Switching and/or combining waveforms contribute to increasing the global SCS responders rate.
Collapse
Affiliation(s)
- Philippe Rigoard
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France; Department of Neuro-Spine & Neuromodulation, Poitiers University Hospital, Poitiers, France; Pprime Institute UPR 3346, CNRS, ISAE-ENSMA, University of Poitiers, Chasseneuil-du-Poitou, France
| | - Amine Ounajim
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France
| | - Maarten Moens
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Brussels, Belgium; STIMULUS research group, Vrije Universiteit Brussel, Brussels, Belgium; Department of Radiology, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Lisa Goudman
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, Brussels, Belgium; STIMULUS research group, Vrije Universiteit Brussel, Brussels, Belgium; Research Foundation-Flanders (FWO), Brussels, Belgium
| | - Manuel Roulaud
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France
| | - Bertille Lorgeoux
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France
| | - Sandrine Baron
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France
| | - Kévin Nivole
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France
| | - Mathilde Many
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France
| | - Lucie Lampert
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France
| | - Romain David
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France; Department of Physical and Rehabilitation Medicine Unit, Poitiers University Hospital, University of Poitiers, Poitiers, France
| | - Maxime Billot
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, Poitiers, France
| |
Collapse
|
32
|
Staats PS, Taylor RS, Gilligan C, Sheth S, Patel KV, Duarte RV, Eldabe S. Limitations of the Cochrane review of spinal cord stimulation for low back pain. Pain Pract 2023; 23:868-872. [PMID: 37427805 DOI: 10.1111/papr.13263] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/05/2023] [Accepted: 06/18/2023] [Indexed: 07/11/2023]
Affiliation(s)
- Peter S Staats
- National Spine and Pain Centers, Shrewsbury, New Jersey, USA
| | - Rod S Taylor
- MRC/CSO Social and Public Health Sciences Unit & Robertson Centre for Biostatistics, Institute of Health and Well Being, University of Glasgow, Glasgow, UK
| | - Christopher Gilligan
- Division of Pain Medicine, Brigham and Women's Hospital Harvard Medical School, Boston, Massachusetts, USA
| | - Samir Sheth
- Sutter Health System, Roseville, California, USA
| | - Kiran V Patel
- The Spine & Pain Institute of New York, New York, New York, USA
- Department of Anesthesiology, NYU Langone Medical Center, New York, New York, USA
| | - Rui V Duarte
- Saluda Medical Pty Ltd., Artarmon, New South Wales, Australia
- Liverpool Reviews and Implementation Group, Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Sam Eldabe
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| |
Collapse
|
33
|
Duarte RV, Bentley A, Soliday N, Leitner A, Gulve A, Staats PS, Sayed D, Falowski SM, Hunter CW, Taylor RS. Cost-utility Analysis of Evoke Closed-loop Spinal Cord Stimulation for Chronic Back and Leg Pain. Clin J Pain 2023; 39:551-559. [PMID: 37440335 PMCID: PMC10498882 DOI: 10.1097/ajp.0000000000001146] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023]
Abstract
OBJECTIVES The effectiveness of Evoke closed-loop spinal cord stimulation (CL-SCS), a novel modality of neurostimulation, has been demonstrated in a randomized controlled trial (RCT). The objective of this cost-utility analysis was to develop a de novo economic model to estimate the cost-effectiveness of Evoke CL-SCS when compared with open-loop SCS (OL-SCS) for the management of chronic back and leg pain. METHODS A decision tree followed by a Markov model was used to estimate the costs and outcomes of Evoke CL-SCS versus OL-SCS over a 15-year time horizon from the UK National Health Service perspective. A "high-responder" health state was included to reflect improved levels of SCS pain reduction recently reported. Results are expressed as incremental cost per quality-adjusted life year (QALY). Deterministic and probabilistic sensitivity analysis (PSA) was conducted to assess uncertainty in the model inputs. RESULTS Evoke CL-SCS was estimated to be the dominant treatment strategy at ~5 years postimplant (ie, it generates more QALYs while cost saving compared with OL-SCS). Probabilistic sensitivity analysis showed that Evoke CL-SCS has a 92% likelihood of being cost-effective at a willingness to pay threshold of £20,000/QALY. Results were robust across a wide range of scenario and sensitivity analyses. DISCUSSION The results indicate a strong economic case for the use of Evoke CL-SCS in the management of chronic back and leg pain with or without prior spinal surgery with dominance observed at ~5 years.
Collapse
Affiliation(s)
- Rui V. Duarte
- Department of Health Data Science, University of Liverpool, Liverpool, UK
- Saluda Medical Pty Ltd., Artarmon, NSW, Australia
| | | | | | | | - Ashish Gulve
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | | | - Dawood Sayed
- The University of Kansas Health System, Kansas City, KS
| | | | | | - Rod S. Taylor
- MRC/CSO Social and Public Health Sciences Unit & Robertson Centre for Biostatistics, Institute of Health and Well Being, University of Glasgow, Glasgow, UK
| |
Collapse
|
34
|
Ege E, Briggi D, Mach S, Huh BK, Javed S. Dorsal root ganglion stimulation for chemotherapy-induced peripheral neuropathy. Pain Pract 2023; 23:793-799. [PMID: 37260046 DOI: 10.1111/papr.13259] [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: 04/15/2023] [Accepted: 05/18/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Chemotherapy-induced peripheral neuropathy (CIPN) is a common consequence of cancer treatment that can be persistent and difficult to manage. Dorsal root ganglion stimulation (DRG-S) is a recently introduced but understudied treatment modality. This study explored the effect of DRG-S on pain and symptom burden associated with CIPN. METHODS Patients with CIPN who underwent a DRG-S trial between January 2017 and August 2022 were identified through chart review after IRB approval was obtained. Demographic data, procedure details, pre-and postoperative scores, including the Numerical Rating Scale (NRS) and Edmonton Symptom Assessment System (ESAS), and duration of follow-up were recorded. Statistical analysis included descriptive statistics and paired t-tests to compare pre-and postoperative scores. RESULTS Nine patients with an even mix of solid and hematologic malignancies underwent DRG-S trial and had a statistically significant decrease in NRS scores, with a mean reduction of 2.3 in their average pain (p = 0.014), 2.6 in worst pain (p = 0.023), and 2.1 in least pain (p = 0.018). Eight patients (88.9%) underwent permanent DRG-S implantation. Mean NRS scores remained lower than preoperative baselines through the first year of follow-up. Statistically significant reductions were noted at 3 months in average (2.1, p = 0.006) and least pain scores (1.9, p = 0.045), which further decreased after 6-12 months (average: 3.6, p = 0.049; least: 3.4, p = 0.023). Only the pain component of ESAS scores showed a significant reduction with DRG-S (2.0, p = 0.021). All patients endorsed improved sensation, 75% decreased their pain medication usage, and 37.5% reported complete pain relief by 2 years. CONCLUSION Dorsal root ganglion stimulation can be an effective treatment for pain related to CIPN and deserves further investigation.
Collapse
Affiliation(s)
- Eliana Ege
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, Texas, USA
| | - Daniel Briggi
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, Texas, USA
| | - Steven Mach
- Department of Pain Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Billy K Huh
- Department of Pain Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Saba Javed
- Department of Pain Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
35
|
Leitner A, Hanson E, Soliday N, Staats P, Levy R, Pope J, Kallewaard JW, Doleys D, Li S, Weisbein J, Amirdelfan K, Poree L. Real World Clinical Utility of Neurophysiological Measurement Utilizing Closed-Loop Spinal Cord Stimulation in a Chronic Pain Population: The ECAP Study Protocol. J Pain Res 2023; 16:2497-2507. [PMID: 37497371 PMCID: PMC10368120 DOI: 10.2147/jpr.s411927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023] Open
Abstract
Background Spinal cord stimulation (SCS) is an established chronic pain treatment, but the effectiveness of traditional, open-loop paradigms has been plagued by variable sustainability in a real-world setting. A new approach, utilizing evoked compound action potential (ECAP) controlled closed-loop (CL) SCS, continuously monitors spinal cord activation and automatically adjusts the stimulation amplitude of every pulse, maintaining stimulation at the prescribed ECAP level through this continual feedback mechanism. Recent studies demonstrated the long-term safety and efficacy of ECAP-controlled CL-SCS. Here, we report the design of a prospective, multicenter, single-arm feasibility study to characterize clinical outcomes in a real-world chronic pain population utilizing ECAP-controlled CL-SCS. Objective neurophysiological measurements such as device performance and patient therapy compliance, will be analyzed against baseline biopsychosocial assessments, to explore the clinical utility of these objective physiologic biomarkers in patient phenotyping. Methods This study will enroll up to 300 subjects with chronic, intractable trunk and/or limb pain in up to 25 United States investigation sites. Subjects meeting eligibility criteria will undergo a trial procedure and a permanent implant following a successful trial. Neurophysiological measurements (measured in-clinic and continuously during home use) and clinical outcomes including pain, quality-of-life, psychological, emotional, and functional assessments will be collected at baseline, trial end, and up to 24-months post-implantation. Discussion Associations between objective neurophysiological data, clinical evaluation and patient-reported outcomes may have important clinical and scientific implications. They may provide novel insights about the chronic pain pathophysiology, its modulation during CL-SCS, and identification of pain phenotypes and/or mechanisms associated with treatment response during SCS trials and long-term therapy. Data from the ECAP study could lead to improvements in diagnosis, assessment, patient identification and management of chronic pain. It could also provide the foundation for development of a new SCS treatment approach customized by the patient's pain phenotype, unique neurophysiology, and disease severity.
Collapse
Affiliation(s)
- Angela Leitner
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Erin Hanson
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Nicole Soliday
- Saluda Medical Pty Ltd, Artarmon, New South Wales, Australia
| | - Peter Staats
- National Spine and Pain Centers, Shrewsbury, NJ, USA
| | - Robert Levy
- Departments of Neurosurgery and Clinical Research, Anesthesia Pain Care Consultants, Tamarac, FL, USA
| | - Jason Pope
- Evolve Restorative Center, Santa Rosa, CA, USA
| | - Jan W Kallewaard
- Department of Anaesthesiology and Pain Management, Rijnstate Hospital, Arnhem, the Netherlands
- Department of Anesthesiology and Pain Medicine, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Daniel Doleys
- Pain and Rehabilitation Institute, Birmingham, AL, USA
| | - Sean Li
- National Spine and Pain Centers, Shrewsbury, NJ, USA
| | | | | | - Lawrence Poree
- Department of Anesthesia and Perioperative Care, University of California at San Francisco, San Francisco, CA, USA
| |
Collapse
|
36
|
Sarica C, Zemmar A, Yousefi O, Yang AC, Uzuner A, Sheng Z, Santyr B, Samuel N, Colditz M, Vetkas A, Germann J, Cheyuo C, Sabahi M, Jani RN, Darmani G, Yamamoto K, Aguirre-Padilla DH, Neimat JS, Kalia SK, Chen R, Fasano AA, Lozano AM. Spinal Cord Stimulation for Parkinson's Disease: A Systematic Review and Meta-Analysis of Pain and Motor Outcomes. Stereotact Funct Neurosurg 2023; 101:244-253. [PMID: 37429256 PMCID: PMC10614495 DOI: 10.1159/000531089] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/03/2023] [Indexed: 07/12/2023]
Abstract
BACKGROUND Spinal cord stimulation (SCS) has been investigated as a potential therapeutic option for managing refractory symptoms in patients with Parkinson's disease (PD). OBJECTIVE This systematic review and meta-analysis aimed to evaluate the safety and efficacy of SCS in PD. METHOD A comprehensive literature search was conducted on PubMed and Web of Science to identify SCS studies reporting Unified Parkinson Disease Rating Scale-III (UPDRS-III) or Visual Analogue Scale (VAS) score changes in PD cohorts with at least 3 patients and a follow-up period of at least 1 month. Treatment effect was measured as the mean change in outcome scores and analyzed using an inverse variance random-effects model. The risk of bias was assessed using the Newcastle-Ottawa Scale and funnel plots. RESULTS A total of 11 studies comprising 76 patients were included. Nine studies involving 72 patients reported an estimated decrease of 4.43 points (95% confidence interval [CI]: 2.11; 6.75, p < 0.01) in UPDRS-III score, equivalent to a 14% reduction. The axial subscores in 48 patients decreased by 2.35 points (95% CI: 1.26; 3.45, p < 0.01, 20% reduction). The pooled effect size of five studies on back and leg pain VAS scores was calculated as 4.38 (95% CI: 2.67; 6.09, p < 0.001), equivalent to a 59% reduction. CONCLUSIONS Our analysis suggests that SCS may provide significant motor and pain benefits for patients with PD, although the results should be interpreted with caution due to several potential limitations including study heterogeneity, open-label designs, small sample sizes, and the possibility of publication bias. Further research using larger sample sizes and placebo-/sham-controlled designs is needed to confirm effectiveness.
Collapse
Affiliation(s)
- Can Sarica
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada,
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada,
| | - Ajmal Zemmar
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurosurgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Omid Yousefi
- Trauma Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Andrew C Yang
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Ayse Uzuner
- Department of Neurosurgery, Kocaeli University, Kocaeli, Turkey
| | - Zhiyuan Sheng
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Brendan Santyr
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Nardin Samuel
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Michael Colditz
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Artur Vetkas
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Jürgen Germann
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Cletus Cheyuo
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Mohammadmahdi Sabahi
- Department of Neurosurgery, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Raja Niranjan Jani
- Department of Neurosurgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Ghazaleh Darmani
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Kazuaki Yamamoto
- Functional Neurosurgery Center, Shonan Fujisawa Tokushukai Hospital, Fujisawa, Japan
| | | | - Joseph S Neimat
- Department of Neurosurgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Suneil K Kalia
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, Ontario, Canada
| | - Robert Chen
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Edmond J. Safra Program in Parkinson's Disease Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital and Division of Neurology, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Alfonso A Fasano
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Center for Advancing Neurotechnological Innovation to Application (CRANIA), Toronto, Ontario, Canada
- Edmond J. Safra Program in Parkinson's Disease Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital and Division of Neurology, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Andres M Lozano
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
37
|
Russo MA, Bhatia A, Hayek S, Doshi T, Eldabe S, Huygen F, Levy RM. Problems With O'Connell et al, "Implanted Spinal Neuromodulation Interventions for Chronic Pain in Adults" (Cochrane Review). Neuromodulation 2023; 26:897-904. [PMID: 37029022 PMCID: PMC10330605 DOI: 10.1016/j.neurom.2023.03.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/05/2023] [Indexed: 04/09/2023]
Affiliation(s)
- Marc A Russo
- Hunter Pain Specialists, Broadmeadow, New South Wales, Australia; Genesis Research Services, Broadmeadow, New South Wales, Australia; University of Newcastle, School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, Callaghan, New South Wales, Australia.
| | - Anuj Bhatia
- Department of Anesthesiology, University of Toronto, Toronto, Ontario, Canada
| | - Salim Hayek
- Division of Pain Medicine, University Hospitals, Cleveland Medical Center, Cleveland, OH, USA
| | - Tina Doshi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sam Eldabe
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Frank Huygen
- Center of Pain Medicine Erasmus Medical Center, Rotterdam, The Netherlands; Center of Pain Medicine University Medical Center Utrecht, Utrecht, The Netherlands
| | - Robert M Levy
- International Neuromodulation Society, Neuromodulation: Technology at the Neural Interface, San Francisco, CA, USA
| |
Collapse
|
38
|
Ferraro MC, Cashin AG, Wand BM, Smart KM, Berryman C, Marston L, Moseley GL, McAuley JH, O'Connell NE. Interventions for treating pain and disability in adults with complex regional pain syndrome- an overview of systematic reviews. Cochrane Database Syst Rev 2023; 6:CD009416. [PMID: 37306570 PMCID: PMC10259367 DOI: 10.1002/14651858.cd009416.pub3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
BACKGROUND Complex regional pain syndrome (CRPS) is a chronic pain condition that usually occurs in a limb following trauma or surgery. It is characterised by persisting pain that is disproportionate in magnitude or duration to the typical course of pain after similar injury. There is currently no consensus regarding the optimal management of CRPS, although a broad range of interventions have been described and are commonly used. This is the first update of the original Cochrane review published in Issue 4, 2013. OBJECTIVES To summarise the evidence from Cochrane and non-Cochrane systematic reviews of the efficacy, effectiveness, and safety of any intervention used to reduce pain, disability, or both, in adults with CRPS. METHODS We identified Cochrane reviews and non-Cochrane reviews through a systematic search of Ovid MEDLINE, Ovid Embase, Cochrane Database of Systematic Reviews, CINAHL, PEDro, LILACS and Epistemonikos from inception to October 2022, with no language restrictions. We included systematic reviews of randomised controlled trials that included adults (≥18 years) diagnosed with CRPS, using any diagnostic criteria. Two overview authors independently assessed eligibility, extracted data, and assessed the quality of the reviews and certainty of the evidence using the AMSTAR 2 and GRADE tools respectively. We extracted data for the primary outcomes pain, disability and adverse events, and the secondary outcomes quality of life, emotional well-being, and participants' ratings of satisfaction or improvement with treatment. MAIN RESULTS: We included six Cochrane and 13 non-Cochrane systematic reviews in the previous version of this overview and five Cochrane and 12 non-Cochrane reviews in the current version. Using the AMSTAR 2 tool, we judged Cochrane reviews to have higher methodological quality than non-Cochrane reviews. The studies in the included reviews were typically small and mostly at high risk of bias or of low methodological quality. We found no high-certainty evidence for any comparison. There was low-certainty evidence that bisphosphonates may reduce pain intensity post-intervention (standardised mean difference (SMD) -2.6, 95% confidence interval (CI) -1.8 to -3.4, P = 0.001; I2 = 81%; 4 trials, n = 181) and moderate-certainty evidence that they are probably associated with increased adverse events of any nature (risk ratio (RR) 2.10, 95% CI 1.27 to 3.47; number needed to treat for an additional harmful outcome (NNTH) 4.6, 95% CI 2.4 to 168.0; 4 trials, n = 181). There was moderate-certainty evidence that lidocaine local anaesthetic sympathetic blockade probably does not reduce pain intensity compared with placebo, and low-certainty evidence that it may not reduce pain intensity compared with ultrasound of the stellate ganglion. No effect size was reported for either comparison. There was low-certainty evidence that topical dimethyl sulfoxide may not reduce pain intensity compared with oral N-acetylcysteine, but no effect size was reported. There was low-certainty evidence that continuous bupivacaine brachial plexus block may reduce pain intensity compared with continuous bupivacaine stellate ganglion block, but no effect size was reported. For a wide range of other commonly used interventions, the certainty in the evidence was very low and provides insufficient evidence to either support or refute their use. Comparisons with low- and very low-certainty evidence should be treated with substantial caution. We did not identify any RCT evidence for routinely used pharmacological interventions for CRPS such as tricyclic antidepressants or opioids. AUTHORS' CONCLUSIONS Despite a considerable increase in included evidence compared with the previous version of this overview, we identified no high-certainty evidence for the effectiveness of any therapy for CRPS. Until larger, high-quality trials are undertaken, formulating an evidence-based approach to managing CRPS will remain difficult. Current non-Cochrane systematic reviews of interventions for CRPS are of low methodological quality and should not be relied upon to provide an accurate and comprehensive summary of the evidence.
Collapse
Affiliation(s)
- Michael C Ferraro
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
- School of Health Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Aidan G Cashin
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
- School of Health Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Benedict M Wand
- The School of Health Sciences and Physiotherapy, The University of Notre Dame Australia, Fremantle, Australia
| | - Keith M Smart
- UCD School of Public Health, Physiotherapy and Sports Science, University College Dublin, Dublin, Ireland
- Physiotherapy Department, St Vincent's University Hospital, Dublin, Ireland
| | - Carolyn Berryman
- IIMPACT in Health, University of South Australia, Kaurna Country, Adelaide, South Australia, Australia
- School of Biomedicine, The University of Adelaide, Kaurna Country, Adelaide, Australia
| | - Louise Marston
- Department of Primary Care and Population Health, University College London, London, UK
| | - G Lorimer Moseley
- IIMPACT in Health, University of South Australia, Kaurna Country, Adelaide, South Australia, Australia
| | - James H McAuley
- Centre for Pain IMPACT, Neuroscience Research Australia, Sydney, Australia
- School of Health Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Neil E O'Connell
- Department of Health Sciences, Centre for Health and Wellbeing Across the Lifecourse, Brunel University London, Uxbridge, UK
| |
Collapse
|
39
|
Duarte RV, Nevitt S, Copley S, Maden M, de Vos CC, Taylor RS, Eldabe S. Response to Comment on Duarte et al. Systematic Review and Network Meta-analysis of Neurostimulation for Painful Diabetic Neuropathy. Diabetes Care 2022;45:2466-2475. Diabetes Care 2023; 46:e113-e114. [PMID: 37185686 DOI: 10.2337/dci23-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Affiliation(s)
- Rui V Duarte
- 1Liverpool Reviews and Implementation Group, University of Liverpool, Liverpool, U.K
- 2Saluda Medical Pty. Ltd., Artarmon, New South Wales, Australia
| | - Sarah Nevitt
- 1Liverpool Reviews and Implementation Group, University of Liverpool, Liverpool, U.K
| | - Sue Copley
- 3Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, U.K
| | - Michelle Maden
- 1Liverpool Reviews and Implementation Group, University of Liverpool, Liverpool, U.K
| | - Cecile C de Vos
- 4Department of Neurology and Neurosurgery, Medisch Spectrum Twente, Enschede, the Netherlands
- 5Centre for Pain Medicine, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Rod S Taylor
- 6Medical Research Council/Chief Scientist Office Social and Public Health Sciences Unit and Robertson Centre for Biostatistics, Institute of Health and Well Being, University of Glasgow, Glasgow, U.K
| | - Sam Eldabe
- 3Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, U.K
| |
Collapse
|
40
|
Enders J, Elliott D, Wright DE. Emerging Nonpharmacologic Interventions to Treat Diabetic Peripheral Neuropathy. Antioxid Redox Signal 2023; 38:989-1000. [PMID: 36503268 PMCID: PMC10402707 DOI: 10.1089/ars.2022.0158] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 12/14/2022]
Abstract
Significance: Diabetic peripheral neuropathy (DPN), a complication of metabolic syndrome, type I and type II diabetes, leads to sensory changes that include slow nerve conduction, nerve degeneration, loss of sensation, pain, and gate disturbances. These complications remain largely untreatable, although tight glycemic control can prevent neuropathy progression. Nonpharmacologic approaches remain the most impactful to date, but additional advances in treatment approaches are needed. Recent Advances: This review highlights several emerging interventions, including a focus on dietary interventions and physical activity, that continue to show promise for treating DPN. We provide an overview of our current understanding of how exercise can improve aspects of DPN. We also highlight new studies in which a ketogenic diet has been used as an intervention to prevent and reverse DPN. Critical Issues: Both exercise and consuming a ketogenic diet induce systemic and cellular changes that collectively improve complications associated with DPN. Both interventions may involve similar signaling pathways and benefits but also impact DPN through unique mechanisms. Future Directions: These lifestyle interventions are critically important as personalized medicine approaches will likely be needed to identify specific subsets of neuropathy symptoms and deficits in patients, and determine the most impactful treatment. Overall, these two interventions have the potential to provide meaningful relief for patients with DPN and provide new avenues to identify new therapeutic targets.
Collapse
Affiliation(s)
- Jonathan Enders
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Daniel Elliott
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Douglas E. Wright
- Department of Anesthesiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
41
|
Sadeghirad B, Wang X, Busse JW. Comment on Duarte et al. Systematic Review and Network Meta-analysis of Neurostimulation for Painful Diabetic Neuropathy. Diabetes Care 2022;45:2466-2475. Diabetes Care 2023; 46:e112. [PMID: 37185685 DOI: 10.2337/dc22-2449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Affiliation(s)
- Behnam Sadeghirad
- 1Department of Anesthesia, McMaster University, Hamilton, Ontario, Canada
- 2Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
- 3Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada
| | - Xiaoqin Wang
- 3Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada
| | - Jason W Busse
- 1Department of Anesthesia, McMaster University, Hamilton, Ontario, Canada
- 2Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
- 3Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
42
|
Shanthanna H, Eldabe S, Provenzano DA, Chang Y, Adams D, Kashir I, Goel A, Tian C, Couban RJ, Levit T, Hagedorn JM, Narouze S. Role of patient selection and trial stimulation for spinal cord stimulation therapy for chronic non-cancer pain: a comprehensive narrative review. Reg Anesth Pain Med 2023; 48:251-272. [PMID: 37001887 DOI: 10.1136/rapm-2022-103820] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/22/2022] [Indexed: 04/03/2023]
Abstract
Background/importancePatient selection for spinal cord stimulation (SCS) therapy is crucial and is traditionally performed with clinical selection followed by a screening trial. The factors influencing patient selection and the importance of trialing have not been systematically evaluated.ObjectiveWe report a narrative review conducted to synthesize evidence regarding patient selection and the role of SCS trials.Evidence reviewMedline, EMBASE and Cochrane databases were searched for reports (any design) of SCS in adult patients, from their inception until March 30, 2022. Study selection and data extraction were carried out using DistillerSR. Data were organized into tables and narrative summaries, categorized by study design. Importance of patient variables and trialing was considered by looking at their influence on the long-term therapy success.FindingsAmong 7321 citations, 201 reports consisting of 60 systematic reviews, 36 randomized controlled trials (RCTs), 41 observational studies (OSs), 51 registry-based reports, and 13 case reports on complications during trialing were included. Based on RCTs and OSs, the median trial success rate was 72% and 82%, and therapy success was 65% and 61% at 12 months, respectively. Although several psychological and non-psychological determinants have been investigated, studies do not report a consistent approach to patient selection. Among psychological factors, untreated depression was associated with poor long-term outcomes, but the effect of others was inconsistent. Most RCTs except for chronic angina involved trialing and only one RCT compared patient selection with or without trial. The median (range) trial duration was 10 (0–30) and 7 (0–56) days among RCTs and OSs, respectively.ConclusionsDue to lack of a consistent approach to identify responders for SCS therapy, trialing complements patient selection to exclude patients who do not find the therapy helpful and/or intolerant of the SCS system. However, more rigorous and large studies are necessary to better evaluate its role.
Collapse
Affiliation(s)
| | - Sam Eldabe
- James Cook University Hospital, Middlesbrough, UK
| | | | - Yaping Chang
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Daniel Adams
- Center for Pain Medicine, Summa Western Reserve Hospital, Cuyahoga Falls, Ohio, USA
| | - Imad Kashir
- University of Waterloo, Waterloo, Ontario, Canada
| | - Akash Goel
- Anesthesiology & Pain Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Chenchen Tian
- Anesthesiology & Pain Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | - Tal Levit
- Michael G DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan M Hagedorn
- Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Samer Narouze
- Center for Pain Medicine, Summa Western Reserve Hospital, Cuyahoga Falls, Ohio, USA
| |
Collapse
|
43
|
Abstract
BACKGROUND Spinal cord stimulation (SCS) is a surgical intervention used to treat persistent low back pain. SCS is thought to modulate pain by sending electrical signals via implanted electrodes into the spinal cord. The long term benefits and harms of SCS for people with low back pain are uncertain. OBJECTIVES To assess the effects, including benefits and harms, of SCS for people with low back pain. SEARCH METHODS On 10 June 2022, we searched CENTRAL, MEDLINE, Embase, and one other database for published trials. We also searched three clinical trials registers for ongoing trials. SELECTION CRITERIA We included all randomised controlled trials and cross-over trials comparing SCS with placebo or no treatment for low back pain. The primary comparison was SCS versus placebo, at the longest time point measured in the trials. Major outcomes were mean low back pain intensity, function, health-related quality of life, global assessment of efficacy, withdrawals due to adverse events, adverse events, and serious adverse events. Our primary time point was long-term follow-up (≥ 12 months). DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included 13 studies with 699 participants: 55% of participants were female; mean age ranged from 47 to 59 years; and all participants had chronic low back pain with mean duration of symptoms ranging from five to 12 years. Ten cross-over trials compared SCS with placebo. Three parallel-group trials assessed the addition of SCS to medical management. Most studies were at risk of performance and detection bias from inadequate blinding and selective reporting bias. The placebo-controlled trials had other important biases, including lack of accounting for period and carryover effects. Two of the three parallel trials assessing SCS as an addition to medical management were at risk of attrition bias, and all three had substantial cross-over to the SCS group for time points beyond six months. In the parallel-group trials, we considered the lack of placebo control to be an important source of bias. None of our included studies evaluated the impact of SCS on mean low back pain intensity in the long term (≥ 12 months). The studies most often assessed outcomes in the immediate term (less than one month). At six months, the only available evidence was from a single cross-over trial (50 participants). There was moderate-certainty evidence that SCS probably does not improve back or leg pain, function, or quality of life compared with placebo. Pain was 61 points (on a 0- to 100-point scale, 0 = no pain) at six months with placebo, and 4 points better (8.2 points better to 0.2 points worse) with SCS. Function was 35.4 points (on a 0- to 100-point scale, 0 = no disability or best function) at six months with placebo, and 1.3 points better (3.9 points better to 1.3 points worse) with SCS. Health-related quality of life was 0.44 points out of 1 (0 to 1 index, 0 = worst quality of life) at six months with placebo, and 0.04 points better (0.16 points better to 0.08 points worse) with SCS. In that same study, nine participants (18%) experienced adverse events and four (8%) required revision surgery. Serious adverse events with SCS included infections, neurological damage, and lead migration requiring repeated surgery. We could not provide effect estimates of the relative risks as events were not reported for the placebo period. In parallel trials assessing SCS as an addition to medical management, it is uncertain whether, in the medium or long term, SCS can reduce low back pain, leg pain, or health-related quality of life, or if it increases the number of people reporting a 50% improvement or better, because the certainty of the evidence was very low. Low-certainty evidence suggests that adding SCS to medical management may slightly improve function and slightly reduce opioid use. In the medium term, mean function (0- to 100-point scale; lower is better) was 16.2 points better with the addition of SCS to medical management compared with medical management alone (95% confidence interval (CI) 19.4 points better to 13.0 points better; I2 = 95%; 3 studies, 430 participants; low-certainty evidence). The number of participants reporting opioid medicine use was 15% lower with the addition of SCS to medical management (95% CI 27% lower to 0% lower; I2 = 0%; 2 studies, 290 participants; low-certainty evidence). Adverse events with SCS were poorly reported but included infection and lead migration. One study found that, at 24 months, 13 of 42 people (31%) receiving SCS required revision surgery. It is uncertain to what extent the addition of SCS to medical management increases the risk of withdrawals due to adverse events, adverse events, or serious adverse events, because the certainty of the evidence was very low. AUTHORS' CONCLUSIONS Data in this review do not support the use of SCS to manage low back pain outside a clinical trial. Current evidence suggests SCS probably does not have sustained clinical benefits that would outweigh the costs and risks of this surgical intervention.
Collapse
Affiliation(s)
- Adrian C Traeger
- Institute for Musculoskeletal Health, The University of Sydney and Sydney Local Health District, Sydney, Australia
- School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Stephen E Gilbert
- Institute for Musculoskeletal Health, The University of Sydney and Sydney Local Health District, Sydney, Australia
- School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Ian A Harris
- Institute for Musculoskeletal Health, The University of Sydney and Sydney Local Health District, Sydney, Australia
- South West Sydney Clinical School, University of New South Wales, Liverpool, Australia
| | - Christopher G Maher
- Institute for Musculoskeletal Health, The University of Sydney and Sydney Local Health District, Sydney, Australia
- School of Public Health, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| |
Collapse
|
44
|
Taylor RS, Eldabe S. Placebo (Sham) Controlled Trials of Spinal Cord Stimulation. Neuromodulation 2023; 26:474-475. [PMID: 36526546 DOI: 10.1016/j.neurom.2022.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/21/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Rod S Taylor
- MRC/CSO Social and Public Health Sciences Unit & Robertson Centre for Biostatistics, Institute of Health and Well Being, University of Glasgow, Glasgow, UK
| | - Sam Eldabe
- Department of Pain Medicine, the James Cook University Hospital, Middlesbrough, UK.
| |
Collapse
|
45
|
Witjes B, Baillet S, Roy M, Oostenveld R, Huygen FJPM, de Vos CC. Heterogeneous Cortical Effects of Spinal Cord Stimulation. Neuromodulation 2023:S1094-7159(22)01405-2. [PMID: 36631377 DOI: 10.1016/j.neurom.2022.12.005] [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: 08/10/2022] [Revised: 11/16/2022] [Accepted: 12/09/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVES The understanding of the cortical effects of spinal cord stimulation (SCS) remains limited. Multiple studies have investigated the effects of SCS in resting-state electroencephalography. However, owing to the large variation in reported outcomes, we aimed to describe the differential cortical responses between two types of SCS and between responders and nonresponders using magnetoencephalography (MEG). MATERIALS AND METHODS We conducted 5-minute resting-state MEG recordings in 25 patients with chronic pain with active SCS in three sessions, each after a one-week exposure to tonic, burst, or sham SCS. We extracted six spectral features from the measured neurophysiological signals: the alpha peak frequency; alpha power ratio (power 7-9 Hz/power 9-11 Hz); and average power in the theta (4-7.5 Hz), alpha (8-12.5 Hz), beta (13-30 Hz), and low-gamma (30.5-60 Hz) frequency bands. We compared these features (using nonparametric permutation t-tests) for MEG sensor and cortical map effects across stimulation paradigms, between participants who reported low (< 5, responders) vs high (≥ 5, nonresponders) pain scores, and in three representative participants. RESULTS We found statistically significant (p < 0.05, false discovery rate corrected) increased MEG sensor signal power below 3 Hz in response to burst SCS compared with tonic and sham SCS. We did not find statistically significant differences (all p > 0.05) between the power spectra of responders and nonresponders. Our data did not show statistically significant differences in the spectral features of interest among the three stimulation paradigms or between responders and nonresponders. These results were confirmed by the MEG cortical maps. However, we did identify certain trends in the MEG source maps for all comparisons and several features, with substantial variation across participants. CONCLUSIONS The considerable variation in cortical responses to the various SCS treatment options necessitates studies with sample sizes larger than commonly reported in the field and more personalized treatment plans. Studies with a finer stratification between responders and nonresponders are required to advance the knowledge on SCS treatment effects.
Collapse
Affiliation(s)
- Bart Witjes
- Center for Pain Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - Sylvain Baillet
- Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| | - Mathieu Roy
- Department of Psychology, McGill University, Montreal, Canada
| | - Robert Oostenveld
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands; NatMEG, Karolinska Institutet, Stockholm, Sweden
| | - Frank J P M Huygen
- Center for Pain Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Cecile C de Vos
- Center for Pain Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands; Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| |
Collapse
|
46
|
Noble T, Boone L, El Helou A. The role of virtual reality as adjunctive therapy to spinal cord stimulation in chronic pain: A feasible concept? FRONTIERS IN PAIN RESEARCH 2023; 4:1094125. [PMID: 36923650 PMCID: PMC10009231 DOI: 10.3389/fpain.2023.1094125] [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: 11/09/2022] [Accepted: 01/31/2023] [Indexed: 03/03/2023] Open
Abstract
Spinal cord stimulation and virtual reality therapy are established and promising techniques, respectively, for managing chronic pain, each with its unique advantages and challenges. While each therapy has been the subject of significant research interest, the prospect of combining the two modalities to offer a synergistic effect in chronic pain therapy is still in its infancy. In this narrative review, we assess the state of the field combining virtual reality as an adjunctive therapy to spinal cord stimulation in chronic pain. We also review the broader field of virtual reality therapy for acute and chronic pain, considering evidence related to feasibility in the Canadian healthcare system from cost and patient satisfaction perspectives. While early results show promise, there are unexplored aspects of spinal cord stimulation combined with virtual reality therapy, particularly long-term effects on analgesia, anxiolysis, and implications on the effectiveness and longevity of spinal cord stimulation. The infrastructure for billing virtual reality as a consult service or therapy must also catch up if it is eventually used to supplement spinal cord stimulation for chronic pain.
Collapse
Affiliation(s)
- Timothy Noble
- Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Lyndon Boone
- Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Antonios El Helou
- Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada.,Division of Neurosurgery, Horizon Health Network, Moncton, NB, Canada
| |
Collapse
|
47
|
Dhruva SS, Murillo J, Ameli O, Morin PE, Spencer DL, Redberg RF, Cohen K. Long-term Outcomes in Use of Opioids, Nonpharmacologic Pain Interventions, and Total Costs of Spinal Cord Stimulators Compared With Conventional Medical Therapy for Chronic Pain. JAMA Neurol 2023; 80:18-29. [PMID: 36441532 PMCID: PMC9706399 DOI: 10.1001/jamaneurol.2022.4166] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/09/2022] [Indexed: 11/29/2022]
Abstract
Importance Spinal cord stimulators (SCSs) are increasingly used for the treatment of chronic pain. There is a need for studies with long-term follow-up. Objective To determine the comparative effectiveness and costs of SCSs compared with conventional medical management (CMM) in a large cohort of patients with chronic pain. Design, Setting, and Participants This was a 1:5 propensity-matched retrospective comparative effectiveness research analysis of insured individuals from April 1, 2016, to August 31, 2018. This study used administrative claims data, including longitudinal medical and pharmacy claims, from US commercial and Medicare Advantage enrollees 18 years or older in Optum Labs Data Warehouse. Patients with incident diagnosis codes for failed back surgery syndrome, complex regional pain syndrome, chronic pain syndrome, and other chronic postsurgical back and extremity pain were included in this study. Data were analyzed from February 1, 2021, to August 31, 2022. Exposures SCSs or CMM. Main Outcomes and Measures Surrogate measures for primary chronic pain treatment modalities, including pharmacologic and nonpharmacologic pain interventions (epidural and facet corticosteroid injections, radiofrequency ablation, and spine surgery), as well as total costs. Results In the propensity-matched population of 7560 patients, mean (SD) age was 63.5 (12.5) years, 3080 (40.7%) were male, and 4480 (59.3%) were female. Among matched patients, during the first 12 months, patients treated with SCSs had higher odds of chronic opioid use (adjusted odds ratio [aOR], 1.14; 95% CI, 1.01-1.29) compared with patients treated with CMM but lower odds of epidural and facet corticosteroid injections (aOR, 0.44; 95% CI, 0.39-0.51), radiofrequency ablation (aOR, 0.57; 95% CI, 0.44-0.72), and spine surgery (aOR, 0.72; 95% CI, 0.61-0.85). During months 13 to 24, there was no significant difference in chronic opioid use (aOR, 1.06; 95% CI, 0.94-1.20), epidural and facet corticosteroid injections (aOR, 1.00; 95% CI, 0.87-1.14), radiofrequency ablation (aOR, 0.84; 95% CI, 0.66-1.09), or spine surgery (aOR, 0.91; 95% CI, 0.75-1.09) with SCS use compared with CMM. Overall, 226 of 1260 patients (17.9%) treated with SCS experienced SCS-related complications within 2 years, and 279 of 1260 patients (22.1%) had device revisions and/or removals, which were not always for complications. Total costs of care in the first year were $39 000 higher with SCS than CMM and similar between SCS and CMM in the second year. Conclusions and Relevance In this large, real-world, comparative effectiveness research study comparing SCS and CMM for chronic pain, SCS placement was not associated with a reduction in opioid use or nonpharmacologic pain interventions at 2 years. SCS was associated with higher costs, and SCS-related complications were common.
Collapse
Affiliation(s)
- Sanket S. Dhruva
- University of California, San Francisco School of Medicine, San Francisco
- Philip R. Lee Institute for Health Policy Studies, University of California, San Francisco, San Francisco
- Department of Medicine, San Francisco Veterans Affairs Medical Center, San Francisco, California
| | - Jaime Murillo
- Optum Labs, UnitedHealth Group, Eden Prairie, Minnesota
| | - Omid Ameli
- Optum Center for Research and Innovation
| | | | | | - Rita F. Redberg
- Philip R. Lee Institute for Health Policy Studies, University of California, San Francisco, San Francisco
- Department of Medicine, University of California, San Francisco School of Medicine, San Francisco
| | - Ken Cohen
- Optum Center for Research and Innovation
| |
Collapse
|
48
|
Duarte RV, Nevitt S, Houten R, Brookes M, Bell J, Earle J, Taylor RS, Eldabe S. Spinal Cord Stimulation for Neuropathic Pain in England From 2010 to 2020: A Hospital Episode Statistics Analysis. Neuromodulation 2023; 26:109-114. [PMID: 35396189 DOI: 10.1016/j.neurom.2022.02.229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/24/2022] [Accepted: 02/07/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVES Spinal cord stimulation (SCS) is a recognized intervention for the management of chronic neuropathic pain. The United Kingdom National Institute of Health and Care Excellence has recommended SCS as a management option for chronic neuropathic pain since 2008. The aim of this study is to undertake an assessment of SCS uptake across the National Health Service in England up to 2020. MATERIALS AND METHODS Hospital Episode Statistics were obtained for patients with neuropathic pain potentially eligible for SCS and patients receiving an SCS-related procedure. Data were retrieved nationally and per region from the years 2010-2011 to 2019-2020. RESULTS There were 50,288 adults in England attending secondary care with neuropathic pain in 2010-2011, increasing to 66,376 in 2019-2020. The number of patients with neuropathic pain with an SCS procedure increased on a year-to-year basis until 2018-2019. However, less than 1% of people with neuropathic pain received an SCS device with no evidence of an increase over time when considering the background increase in neuropathic pain prevalence. CONCLUSION Only a small proportion of patients in England with neuropathic pain potentially eligible for SCS receives this intervention. The recommendation for routine use of SCS for management of neuropathic pain has not resulted in an uptake of SCS over the last decade.
Collapse
Affiliation(s)
- Rui V Duarte
- Liverpool Reviews and Implementation Group, Department of Health Data Science, University of Liverpool, Liverpool, UK.
| | - Sarah Nevitt
- Liverpool Reviews and Implementation Group, Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Rachel Houten
- Liverpool Reviews and Implementation Group, Department of Health Data Science, University of Liverpool, Liverpool, UK
| | - Morag Brookes
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Jill Bell
- Patient and Public Involvement Representatives, Middlesbrough, UK
| | - Jenny Earle
- Patient and Public Involvement Representatives, Middlesbrough, UK
| | - Rod S Taylor
- College of Medicine and Health, University of Exeter, Exeter, UK; MRC/CSO Social and Public Health Sciences Unit & Robertson Centre for Biostatistics, Institute of Health and Well Being, University of Glasgow, Glasgow, UK
| | - Sam Eldabe
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| |
Collapse
|
49
|
Opova K, Limousin P, Akram H. Spinal Cord Stimulation for Gait Disorders in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2023; 13:57-70. [PMID: 36683516 PMCID: PMC9912734 DOI: 10.3233/jpd-223284] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 12/22/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Spinal cord stimulation (SCS) is a therapeutic procedure widely used in the management of refractory chronic pain. Evidence from case reports and small descriptive studies has emerged suggesting a role for SCS in patients with gait dysfunction, such as freezing of gait (FoG) and postural imbalance. These are severely debilitating symptoms of advanced Parkinson's disease (PD). OBJECTIVE To establish the current evidence base for the potential application of SCS on gait and balance dysfunction in PD patients. METHODS Three online databases were screened for relevant manuscripts. Two separate searches and four different search strategies were applied to yield relevant results. The main parameters of interest were postural and gait symptoms; secondary outcomes were Quality of Life (QoL) and adverse effects. RESULTS Nineteen studies fulfilled the inclusion criteria. Motor improvements using section III of the Unified Parkinson's Disease Rating Score (UPDRS-III) were available in 13 studies. Measurements to assess FoG reported the following improvements: FoG questionnaires (in 1/19 studies); generalized freezing parameters (2); and walkway/wireless accelerometer measurements (2). Parameters of postural imbalance and falling improved as follows: BBS (1); posture sagittal vertical axis (1); and generalized data on postural instability (8). Two studies reported on adverse effects. QoL was shown to improve as follows: EQ-5D (2); ADL (1); SF-36 (1); BDI-II (1); PDQ-8 (1); HDRS (1); and VAS (5). CONCLUSION SCS may have a therapeutic potential in advanced PD patients suffering from postural and gait-related symptoms. The existing evidence suggests that SCS positively affects patients' QoL with an acceptable safety profile in this patient population.
Collapse
Affiliation(s)
- Karolina Opova
- Unit of Functional Neurosurgery, Queen Square Institute of Neurology, University College London (UCL), London, UK
| | - Patricia Limousin
- Unit of Functional Neurosurgery, Queen Square Institute of Neurology, University College London (UCL), London, UK
| | - Harith Akram
- Unit of Functional Neurosurgery, Queen Square Institute of Neurology, University College London (UCL), London, UK
- Victor Horsley Department of Neurosurgery, the National Hospital for Neurology and Neurosurgery, University College London Hospitals (UCLH), Queen Square, London, UK
| |
Collapse
|
50
|
Eldabe S, Nevitt S, Griffiths S, Gulve A, Thomson S, Baranidharan G, Houten R, Brookes M, Kansal A, Earle J, Bell J, Taylor RS, Duarte RV. Does a Screening Trial for Spinal Cord Stimulation in Patients With Chronic Pain of Neuropathic Origin Have Clinical Utility (TRIAL-STIM)? 36-Month Results From a Randomized Controlled Trial. Neurosurgery 2023; 92:75-82. [PMID: 36226961 PMCID: PMC10158909 DOI: 10.1227/neu.0000000000002165] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/20/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Screening trials before full implantation of a spinal cord stimulation device are recommended by clinical guidelines and regulators, although there is limited evidence for their use. The TRIAL-STIM study showed that a screening trial strategy does not provide superior patient pain outcome at 6-month follow-up compared with not doing a screening trial and that it was not cost-effective. OBJECTIVE To report the long-term follow-up results of the TRIAL-STIM study. METHODS The primary outcome of this pragmatic randomized controlled trial was pain intensity as measured on a numerical rating scale (NRS) and secondary outcomes were the proportion of patients achieving at least 50% and 30% pain relief at 6 months, health-related quality of life, and complication rates. RESULTS Thirty patients allocated to the "Trial Group" (TG) and 36 patients allocated to the "No Trial Group" (NTG) completed outcome assessment at 36-month follow-up. Although there was a reduction in NRS pain and improvements in utility scores from baseline to 36 months in both groups, there was no difference in the primary outcome of pain intensity NRS between TG and NTG (adjusted mean difference: -0.60, 95% CI: -1.83 to 0.63), EuroQol-5 Dimension utility values (adjusted mean difference: -0.02, 95% CI: -0.13 to 0.10), or proportion of pain responders (33% TG vs 31% NTG). No differences were observed between the groups for the likelihood of spinal cord stimulation device explant or reporting an adverse advent up to 36-month follow-up. CONCLUSION The long-term results show no patient outcome benefit in undertaking an SCS screening trial.
Collapse
Affiliation(s)
- Sam Eldabe
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Sarah Nevitt
- Liverpool Reviews and Implementation Group, University of Liverpool, Liverpool, UK
| | - Sara Griffiths
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Ashish Gulve
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Simon Thomson
- Department of Pain Medicine and Neuromodulation, Mid and South Essex University Hospitals, Essex,UK
| | | | - Rachel Houten
- Liverpool Reviews and Implementation Group, University of Liverpool, Liverpool, UK
| | - Morag Brookes
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Anu Kansal
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Jenny Earle
- Patient and Public Involvement Representatives, Middlesbrough, UK
| | - Jill Bell
- Patient and Public Involvement Representatives, Middlesbrough, UK
| | - Rod S. Taylor
- College of Medicine and Health, University of Exeter, Exeter, UK
- MRC/CSO Social and Public Health Sciences Unit & Robertson Centre for Biostatistics, Institute of Health and Well Being, University of Glasgow, Glasgow, UK
| | - Rui V. Duarte
- Liverpool Reviews and Implementation Group, University of Liverpool, Liverpool, UK
| |
Collapse
|