1
|
Finney JN, Levy IR, Chandrasekaran S, Collinger JL, Boninger ML, Gaunt RA, Helm ER, Fisher LE. Techniques to mitigate lead migration for percutaneous trials of cervical spinal cord stimulation. Front Surg 2025; 12:1458572. [PMID: 40230710 PMCID: PMC11994673 DOI: 10.3389/fsurg.2025.1458572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 03/05/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction Epidural spinal cord stimulation (SCS) is a clinical neuromodulation technique that is commonly used to treat neuropathic pain, with patients typically undergoing a one-week percutaneous trial phase before permanent implantation. Traditional SCS involves stimulation of the thoracic spinal cord, but there has been substantial recent interest in cervical SCS to treat upper extremity pain, restore sensation from the missing hand after amputation, or restore motor function to paretic limbs in people with stroke and spinal cord injury. Because of the additional mobility of the neck, as compared to the trunk, lead migration can be a major challenge for cervical SCS, especially during the percutaneous trial phase. The objective of this study was to optimize the implantation procedure of cervical SCS leads to minimize lead migration and increase lead stability during SCS trials. Methods In this study, four subjects underwent percutaneous placement of three SCS leads targeting the cervical spinal segments as part of a clinical trial aiming to restore sensation for people with upper-limb amputation. The leads were maintained for up to 29 days and weekly x-ray imaging was used to measure rostrocaudal and mediolateral lead migration based on bony landmarks. Results and discussion Lead migration was primarily confined to the rostrocaudal axis with the most migration occurring during the first week. Iterative improvements to the implantation procedure were implemented to increase lead stability across subjects. There was a decrease in lead migration for patients who had more rostral placement of the SCS leads. The average migration from the day of lead implant to lead removal was 29.7 mm for Subject 1 (lead placement ranging from T3-T4 to T1-T2), 41.9 mm for Subject 2 (T2-T3 to C7-T1), 1.9 mm for Subject 3 (T1-T2 to C7-T1), and 16.6 mm for Subject 4 (T1-T2 to C7-T1). We found that initial placement of spinal cord stimulator leads in the cervical epidural space as rostral as possible was critical to minimizing subsequent rostrocaudal lead migration.
Collapse
Affiliation(s)
- Jonathan N. Finney
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States
| | - Isaiah R. Levy
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States
- Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA, United States
| | - Santosh Chandrasekaran
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States
- Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA, United States
- Neural Bypass and Brain Computer Interface Lab, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Jennifer L. Collinger
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States
- Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Michael L. Boninger
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States
- Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Robert A. Gaunt
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States
- Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Eric R. Helm
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lee E. Fisher
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States
- Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| |
Collapse
|
2
|
Dalrymple AN, Jones ST, Fallon JB, Shepherd RK, Weber DJ. Overcoming failure: improving acceptance and success of implanted neural interfaces. Bioelectron Med 2025; 11:6. [PMID: 40083033 PMCID: PMC11907899 DOI: 10.1186/s42234-025-00168-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/06/2025] [Indexed: 03/16/2025] Open
Abstract
Implanted neural interfaces are electronic devices that stimulate or record from neurons with the purpose of improving the quality of life of people who suffer from neural injury or disease. Devices have been designed to interact with neurons throughout the body to treat a growing variety of conditions. The development and use of implanted neural interfaces is increasing steadily and has shown great success, with implants lasting for years to decades and improving the health and quality of life of many patient populations. Despite these successes, implanted neural interfaces face a multitude of challenges to remain effective for the lifetime of their users. The devices are comprised of several electronic and mechanical components that each may be susceptible to failure. Furthermore, implanted neural interfaces, like any foreign body, will evoke an immune response. The immune response will differ for implants in the central nervous system and peripheral nervous system, as well as over time, ultimately resulting in encapsulation of the device. This review describes the challenges faced by developers of neural interface systems, particularly devices already in use in humans. The mechanical and technological failure modes of each component of an implant system is described. The acute and chronic reactions to devices in the peripheral and central nervous system and how they affect system performance are depicted. Further, physical challenges such as micro and macro movements are reviewed. The clinical implications of device failures are summarized and a guide for determining the severity of complication was developed and provided. Common methods to diagnose and examine mechanical, technological, and biological failure modes at various stages of development and testing are outlined, with an emphasis on chronic in vivo characterization of implant systems. Finally, this review concludes with an overview of some of the innovative solutions developed to reduce or resolve the challenges faced by implanted neural interface systems.
Collapse
Affiliation(s)
- Ashley N Dalrymple
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
- Department of Physical Medicine and Rehabilitation, University of Utah, Salt Lake City, UT, USA.
- NERVES Lab, University of Utah, Salt Lake City, UT, USA.
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
- NeuroMechatronics Lab, Carnegie Mellon University, Pittsburgh, PA, USA.
| | - Sonny T Jones
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
- NERVES Lab, University of Utah, Salt Lake City, UT, USA
| | - James B Fallon
- Bionics Institute, St. Vincent's Hospital, Melbourne, VIC, Australia
- Medical Bionics Department, University of Melbourne, Melbourne, VIC, Australia
| | - Robert K Shepherd
- Bionics Institute, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Douglas J Weber
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
- NeuroMechatronics Lab, Carnegie Mellon University, Pittsburgh, PA, USA
- Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Chung M, Abd-Elsayed A. Comparative efficacy of closed-loop spinal cord stimulation and dorsal root ganglion stimulation through combination trialing for cancer pain - A retrospective case series. Pain Pract 2025; 25:e70010. [PMID: 39831333 DOI: 10.1111/papr.70010] [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] [Indexed: 01/22/2025]
Abstract
OBJECTIVE To compare the efficacy of closed-loop spinal cord stimulation (CL-SCS) and dorsal root ganglion (DRG) stimulation in managing chronic cancer-related pain. MATERIAL/METHODS A retrospective review was conducted with IRB exemption for four patients with cancer-related pain who underwent combination stimulator trials. Patients were trialed with both CL-SCS and DRG stimulation for 8-10 days, with assessments of pain relief, functional improvement, sleep improvement, pain medication changes, and overall satisfaction. RESULTS All four patients reported significant relief and functional improvement with both CL-SCS and DRG stimulations. CL-SCS provided a range of 12.9-19.6 million adjustments during the trial period. Neural dose was delivered at a median dose ratio of 1.35 and median dose accuracy of 4.9 μV. Patients 1-3 preferred CL-SCS for paresthesia-based stimulation, having the potential to expand pain coverage, full-body MRI compatibility, and real-time automated adjustment features. Patient 4 favored DRG for its ability to provide slightly greater relief, over one aspect of his pain area, despite having comparable coverage with CL-SCS. CONCLUSIONS The consistent and adaptable delivery of CL-SCS may bridge the gap in efficacy that DRG therapy has held over traditional dorsal column stimulation for historically complex and focal conditions that suggests further investigation.
Collapse
Affiliation(s)
- Matthew Chung
- Department of Pain Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Alaa Abd-Elsayed
- Department of Anesthesiology, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
4
|
Kim DC, Kang E, Lee HS, Park YH, Lee B, Kwon JY, Moon J, Lee SE. Challenges in removing an aged spinal cord stimulator: A case study of complete fracture in a 9-year-old S-series paddle lead. Pain Pract 2025; 25:e13429. [PMID: 39440391 DOI: 10.1111/papr.13429] [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] [Indexed: 10/25/2024]
Abstract
INTRODUCTION This case report presents an instance of an S-Series™ slim paddle lead fracturing during extraction, highlighting potential risks associated with the removal of this lead. CASE REPORT A 47-year-old male with complex regional pain syndrome type 2, unresponsive to pharmacotherapy, had undergone the implantation of two spinal cord stimulator (SCS) leads, an Octrode™ cylindrical and an S-series™ slim paddle, using the Epiducer™ system (St Jude Medical) 9 years earlier, with a subsequent intrathecal baclofen pump installed 1 year after SCS. Initially, these interventions stabilized the patient's pain symptoms. However, the diminishing effectiveness of SCS, coupled with a decrease in battery life and increased opioid consumption, necessitated recent surgical procedures. These included the removal and replacement of the implantable pulse generator (IPG) and leads to improve pain management and ensure MRI compatibility. During the removal of the S-series™ slim paddle type lead, complications arose, leading to the retention of an electrode fragment, which necessitated abandoning the replacement of both the IPG and lead. Post-surgical assessments revealed no new neurological impairments, and imaging studies confirmed the stable position of the retained fragment. The patient was discharged with a continued comprehensive pain management plan. CONCLUSION This case highlights the challenges and risks of percutaneous removal of slim paddle type leads, emphasizing the need for careful procedural planning and consideration of surgical options to avoid complications. Further research is needed to evaluate the long-term durability and removal risks of various SCS lead types.
Collapse
Affiliation(s)
- Dong-Chun Kim
- Department of Anesthesiology and Pain Medicine, Haeundae Paik Hospital, Inje University, Busan, South Korea
| | - Eunsu Kang
- Department of Anesthesiology and Pain Medicine, Haeundae Paik Hospital, Inje University, Busan, South Korea
| | - Hyun-Seong Lee
- Department of Anesthesiology and Pain Medicine, Haeundae Paik Hospital, Inje University, Busan, South Korea
| | - Yei Heum Park
- Department of Anesthesiology and Pain Medicine, Haeundae Paik Hospital, Inje University, Busan, South Korea
| | - Byeongcheol Lee
- Department of Anesthesiology and Pain Medicine, Haeundae Paik Hospital, Inje University, Busan, South Korea
| | - Ji Yeon Kwon
- Department of Anesthesiology and Pain Medicine, Haeundae Paik Hospital, Inje University, Busan, South Korea
| | - Junseong Moon
- Department of Anesthesiology and Pain Medicine, Haeundae Paik Hospital, Inje University, Busan, South Korea
| | - Sang Eun Lee
- Department of Anesthesiology and Pain Medicine, Haeundae Paik Hospital, Inje University, Busan, South Korea
| |
Collapse
|
5
|
Waris A, Siraj M, Khan A, Lin J, Asim M, Alhumaydh FA. A Comprehensive Overview of the Current Status and Advancements in Various Treatment Strategies against Epilepsy. ACS Pharmacol Transl Sci 2024; 7:3729-3757. [PMID: 39698272 PMCID: PMC11650742 DOI: 10.1021/acsptsci.4c00494] [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: 08/16/2024] [Revised: 10/10/2024] [Accepted: 10/21/2024] [Indexed: 12/20/2024]
Abstract
Epilepsy affects more than 70 million individuals of all ages worldwide and remains one of the most severe chronic noncommunicable neurological diseases globally. Several neurotransmitters, membrane protein channels, receptors, enzymes, and, more recently noted, various pathways, such as inflammatory and mTORC complexes, play significant roles in the initiation and propagation of seizures. Over the past two decades, significant developments have been made in the diagnosis and treatment of epilepsy. Various pharmacological drugs with diverse mechanisms of action and other treatment options have been developed to control seizures and treat epilepsy. These options include surgical treatment, nanomedicine, gene therapy, natural products, nervous stimulation, a ketogenic diet, gut microbiota, etc., which are in various developmental stages. Despite a plethora of drugs and other treatment options, one-third of affected individuals are resistant to current medications, while the majority of approved drugs have severe side effects, and significant changes can occur, such as pharmacoresistance, effects on cognition, long-term problems, drug interactions, risks of poor adherence, specific effects for certain medications, and psychological complications. Therefore, the development of new drugs and other treatment options that have no or minimal adverse effects is needed to combat this deadly disease. In this Review, we comprehensively summarize and explain all of the treatment options that have been approved or are in developmental stages for epilepsy as well as their status in clinical trials and advancements.
Collapse
Affiliation(s)
- Abdul Waris
- Department
of Biomedical Science, City University of
Hong Kong, 999077 Hong Kong SAR
| | - Muhammad Siraj
- Department
of Biotechnology, Jeonbuk National University−Iksan
Campus, Jeonju 54896, South Korea
| | - Ayyaz Khan
- Department
of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju 54907, South Korea
| | - Junyu Lin
- Department
of Neuroscience, City University of Hong
Kong, 999077 Hong Kong SAR
| | - Muhammad Asim
- Department
of Neuroscience, City University of Hong
Kong, 999077 Hong Kong SAR
| | - Fahad A. Alhumaydh
- Department
of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| |
Collapse
|
6
|
Ting JE, Hooper CA, Dalrymple AN, Weber DJ. Tonic Stimulation of Dorsal Root Ganglion Results in Progressive Decline in Recruitment of Aα/β-Fibers in Rats. Neuromodulation 2024; 27:1347-1359. [PMID: 39046395 PMCID: PMC11625011 DOI: 10.1016/j.neurom.2024.06.498] [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: 03/23/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024]
Abstract
OBJECTIVES In this study, we aimed to characterize the recruitment and maintenance of action potential firing in Aα/β-fibers generated during tonic dorsal root ganglion stimulation (DRGS) applied over a range of clinically relevant stimulation parameters. MATERIALS AND METHODS We delivered electrical stimulation to the L5 dorsal root ganglion and recorded antidromic evoked compound action potentials (ECAPs) in the sciatic nerve during DRGS in Sprague Dawley rats. We measured charge thresholds to elicit ECAPs in Aα/β-fibers during DRGS applied at multiple pulse widths (50, 150, 300, 500 μs) and frequencies (5, 20, 50, 100 Hz). We measured the peak-to-peak amplitudes, latencies, and widths of ECAPs generated during 180 seconds of DRGS, and excitation threshold changes to investigate potential mechanisms of ECAP suppression. RESULTS Tonic DRGS produced ECAPs in Aα/β-fibers at charge thresholds below the motor threshold. Increasing the pulse width of DRGS led to a significant increase in the charge required to elicit ECAPs in Aα/β-fibers, while varying DRGS frequency did not influence ECAP thresholds. Over the course of 180 seconds, ECAP peak-to-peak amplitude decreased progressively in a frequency-dependent manner, where 5- and 100-Hz DRGS resulted in 22% and 87% amplitude reductions, respectively, and ECAP latencies increased from baseline measurements during DRGS at 10, 20, 50, and 100 Hz. Regardless of DRGS frequency, ECAP amplitudes recovered within 120 seconds after turning DRGS off. We determined that ECAP suppression may be attributed to increasing excitation thresholds for individual fibers during DRGS. Following 180 seconds of DRGS, an average of 7.33% increase in stimulation amplitude was required to restore the ECAP to baseline amplitude. CONCLUSIONS DRGS produces a progressive and frequency-dependent reduction in ECAP amplitude that occurs within and above the frequency range used clinically to relieve pain. If DRGS-mediated analgesia relies on Aβ-fiber activation, then the frequency or duty cycle of stimulation should be set to the lowest effective level to maintain sufficient activation of Aβ-fibers.
Collapse
Affiliation(s)
- Jordyn E Ting
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Charli Ann Hooper
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Ashley N Dalrymple
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Department of Physical Medicine and Rehabilitation, University of Utah, Salt Lake City, UT, USA
| | - Douglas J Weber
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Neuroscience Institute, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Graczyk E, Hutchison B, Valle G, Bjanes D, Gates D, Raspopovic S, Gaunt R. Clinical Applications and Future Translation of Somatosensory Neuroprostheses. J Neurosci 2024; 44:e1237242024. [PMID: 39358021 PMCID: PMC11450537 DOI: 10.1523/jneurosci.1237-24.2024] [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: 06/30/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 10/04/2024] Open
Abstract
Somatosensory neuroprostheses restore, replace, or enhance tactile and proprioceptive feedback for people with sensory impairments due to neurological disorders or injury. Somatosensory neuroprostheses typically couple sensor inputs from a wearable device, prosthesis, robotic device, or virtual reality system with electrical stimulation applied to the somatosensory nervous system via noninvasive or implanted interfaces. While prior research has mainly focused on technology development and proof-of-concept studies, recent acceleration of clinical studies in this area demonstrates the translational potential of somatosensory neuroprosthetic systems. In this review, we provide an overview of neurostimulation approaches currently undergoing human testing and summarize recent clinical findings on the perceptual, functional, and psychological impact of somatosensory neuroprostheses. We also cover current work toward the development of advanced stimulation paradigms to produce more natural and informative sensory feedback. Finally, we provide our perspective on the remaining challenges that need to be addressed prior to translation of somatosensory neuroprostheses.
Collapse
Affiliation(s)
- Emily Graczyk
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio 44106
| | - Brianna Hutchison
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106
| | - Giacomo Valle
- Department of Electrical Engineering, Chalmers University of Technology, Goteborg 41296, Sweden
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, Illinois 60637
| | - David Bjanes
- Division of Biology and Biological Engineering and Tianqiao & Chrissy Chen Brain-Machine Interface Center, California Institute of Technology, Pasadena, California 91125
| | - Deanna Gates
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109
- School of Kinesiology, University of Michigan, Ann Arbor, Michigan 48109
| | - Stanisa Raspopovic
- Laboratory for Neuroengineering, Department of Health Sciences and Technology, Institute for Robotics and Intelligent Systems, ETH Zurich, Zurich 8092, Switzerland
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna 1090, Austria
| | - Robert Gaunt
- Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
8
|
Her YF, Churchill RA. Case Report: Rescue of Relapsed Pain in a Patient with Complex Regional Pain Syndrome Type II by Adding Another Dorsal Root Ganglion Lead. Int Med Case Rep J 2024; 17:765-769. [PMID: 39220373 PMCID: PMC11363934 DOI: 10.2147/imcrj.s477303] [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: 05/08/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
We present on a patient with complex regional pain syndrome (CRPS) following ankle surgery. Pain was refractory to both conservative and surgical measures including neurotomies, ankle fusion, hardware removal, and spinal cord stimulation (SCS) trial. A dorsal root ganglion (DRG) stimulation trial with lead placements at L4, L5, and S1 provided significant pain and functional improvement. However, during the implantation, we were able to place only two DRG leads at L4 and L5 and not S1 due to difficulties with advancing the lead to the desired location. Nonetheless, the two DRG leads provided 90% pain relief and 75% functional improvement for 9 months. However, the patient experienced pain symptoms similar to that of pre-implant without a clear trigger after 9 months despite no DRG stimulator hardware malfunction or lead migration. A decision was made to re-try implanting the S1 DRG lead, which was successful and provided significant pain relief.
Collapse
Affiliation(s)
- Yeng F Her
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Robert A Churchill
- Mayo Clinic Alix School of Medicine, Mayo Clinic Hospital, Rochester, MN, 55905, USA
| |
Collapse
|
9
|
Reinders LJ, Luijten JAM, Frankema SPG, Huygen FJPM, de Vos CC. The Effect of Various Spinal Neurostimulation Paradigms on the Supraspinal Somatosensory Evoked Response: A Systematic Review. Neuromodulation 2024; 27:1008-1019. [PMID: 38752944 DOI: 10.1016/j.neurom.2024.04.003] [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: 01/26/2024] [Revised: 03/19/2024] [Accepted: 04/08/2024] [Indexed: 08/09/2024]
Abstract
INTRODUCTION Spinal neurostimulation is a therapy for otherwise intractable chronic pain. Spinal neurostimulation includes stimulation of the spinal cord (SCS), dorsal root ganglion (DRGS), and dorsal root entry zone (DREZS). New paresthesia-free neurostimulation paradigms may rely on different mechanisms of action from those of conventional tonic neurostimulation. The aim of this systematic review is to assess the existing knowledge on the effect of spinal neurostimulation on somatosensory processing in patients with chronic pain. We therefore reviewed the existing literature on the effect of various spinal neurostimulation paradigms on the supraspinal somatosensory evoked response (SER). MATERIALS AND METHODS Multiple scientific data bases were searched for studies that assessed the effect of spinal neurostimulation on the supraspinal SER, evoked by painful or nonpainful peripheral stimuli in patients with chronic pain. We found 205 studies, of which 24 were included. Demographic data, study design, and study outcome were extracted. RESULTS Of the 24 included studies, 23 used electroencephalography to assess the SER; one study used magnetoencephalography. Fifteen studies evaluated tonic SCS; six studies (also) evaluated paresthesia-free paradigms; three studies evaluated the effect of tonic DRGS or DREZS. Sixteen studies used nonpainful stimuli to elicit the SER, 14 observed a decreased SER amplitude. Ten studies used painful stimuli to elicit the SER, yielding mixed results. DISCUSSION The included studies suggest that both paresthesia-based and paresthesia-free spinal neurostimulation paradigms can decrease (part of) the SER elicited by a nonpainful peripheral stimulus. The observed SER amplitude reduction likely is the effect of various spinal and supraspinal mechanisms of spinal neurostimulation that also contribute to pain relief. CONCLUSIONS Spinal neurostimulation modulates the processing of a peripherally applied nonpainful stimulus. For painful stimuli, the results are not conclusive. It is not yet clear whether paresthesia-free neurostimulation affects the SER differently from paresthesia-based neurostimulation.
Collapse
Affiliation(s)
- Laurien J Reinders
- Erasmus University Medical Center, Center for Pain Medicine, Anesthesiology, Rotterdam, The Netherlands.
| | - Janne A M Luijten
- Erasmus University Medical Center, Center for Pain Medicine, Anesthesiology, Rotterdam, The Netherlands
| | - Sander P G Frankema
- Erasmus University Medical Center, Center for Pain Medicine, Anesthesiology, Rotterdam, The Netherlands
| | - Frank J P M Huygen
- Erasmus University Medical Center, Center for Pain Medicine, Anesthesiology, Rotterdam, The Netherlands
| | - Cecile C de Vos
- Erasmus University Medical Center, Center for Pain Medicine, Anesthesiology, Rotterdam, The Netherlands
| |
Collapse
|
10
|
Ladez SR, Liu J, Chen L, Feng B. Computational Modeling of Dorsal Root Ganglion Stimulation: Understanding Pain Suppression Mechanisms. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2024; 2024:1-4. [PMID: 40039770 DOI: 10.1109/embc53108.2024.10782923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
This study aims to advance our mechanistic understanding of electrical stimulation of dorsal root ganglia (DRG) for treating chronic pain. While DRG stimulation has shown moderate clinical success in managing certain types of chronic pain, the underlying neural mechanism remains inconclusive, hindering the further development of the technology to treat a broader range of chronic pain symptoms and benefit a larger patient population. In this study, we conducted computational simulations in the NEURON simulation environment to assess the neuromodulatory effect of DRG stimulation on action potential transmission in Aδ-fiber and C-fiber sensory afferents. Our simulation incorporates Markov-type state models to capture the subtle gating characteristics of voltage-gated sodium channel subtypes, especially NaV1.6, the anatomical distribution of which was revealed by our immunohistological staining on sparsely labeled afferents. Our simulation results indicate that DRG stimulation causes a significant increase in intra-axonal Na+ concentration and a reduction in K+ concentration, collectively disrupting the transaxonal ionic gradients. This disruption resulted in activitydependent conduction slowing, leading to the eventual conduction block in both Aδ- and C-fiber afferents. This research marks a crucial step forward in unraveling the intricate mechanisms underlying DRG stimulation, presenting a framework for the further development of innovative pain modulation strategies that target the DRG.
Collapse
|
11
|
Donati E, Valle G. Neuromorphic hardware for somatosensory neuroprostheses. Nat Commun 2024; 15:556. [PMID: 38228580 PMCID: PMC10791662 DOI: 10.1038/s41467-024-44723-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/03/2024] [Indexed: 01/18/2024] Open
Abstract
In individuals with sensory-motor impairments, missing limb functions can be restored using neuroprosthetic devices that directly interface with the nervous system. However, restoring the natural tactile experience through electrical neural stimulation requires complex encoding strategies. Indeed, they are presently limited in effectively conveying or restoring tactile sensations by bandwidth constraints. Neuromorphic technology, which mimics the natural behavior of neurons and synapses, holds promise for replicating the encoding of natural touch, potentially informing neurostimulation design. In this perspective, we propose that incorporating neuromorphic technologies into neuroprostheses could be an effective approach for developing more natural human-machine interfaces, potentially leading to advancements in device performance, acceptability, and embeddability. We also highlight ongoing challenges and the required actions to facilitate the future integration of these advanced technologies.
Collapse
Affiliation(s)
- Elisa Donati
- Institute of Neuroinformatics, University of Zurich and ETH Zurich, Zurich, Switzerland.
| | - Giacomo Valle
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
12
|
Mons MR, Chapman KB, Terwiel C, Joosten EA, Kallewaard JW. Burst Spinal Cord Stimulation as Compared With L2 Dorsal Root Ganglion Stimulation in Pain Relief for Nonoperated Discogenic Low Back Pain: Analysis of Two Prospective Studies. Neuromodulation 2024; 27:172-177. [PMID: 37191612 DOI: 10.1016/j.neurom.2023.04.464] [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: 11/03/2022] [Revised: 03/26/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Chronic discogenic low back pain (CD-LBP) is caused by degenerated disks marked by neural and vascular ingrowth. Spinal cord stimulation (SCS) has been shown to be effective for pain relief in patients who are not responsive to conventional treatments. Previously, the pain-relieving effect of two variations of SCS has been evaluated in CD-LBP: Burst SCS and L2 dorsal root ganglion stimulation (DRGS). The aim of this study is to compare the effectivity in pain relief and pain experience of Burst SCS with that of conventional L2 DRGS in patients with CD-LBP. MATERIALS AND METHODS Subjects were implanted with either Burst SCS (n = 14) or L2 DRGS with conventional stimulation (n = 15). Patients completed the numeric pain rating score (NRS) for back pain and Oswestry disability index (ODI) and EuroQoL 5D (EQ-5D) questionnaires at baseline, and at three, six, and 12 months after implantation. Data were compared between time points and between groups. RESULTS Both Burst SCS and L2 DRGS significantly decreased NRS, ODI, and EQ-5D scores as compared with baseline. L2 DRGS resulted in significantly lower NRS scores at 12 months and significantly increased EQ-5D scores at six and 12 months. CONCLUSIONS Both L2 DRGS and Burst SCS resulted in reduction of pain and disability, and increased quality of life in patients with CD-LBP. L2 DRGS provided significantly increased pain relief and improvement in quality of life when compared with Burst SCS. CLINICAL TRIAL REGISTRATION The clinical trial registration numbers for the study are NCT03958604 and NL54405.091.15.
Collapse
Affiliation(s)
- Martijn R Mons
- Department of Anesthesiology and Pain Management, University Pain Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands; Department of Translational Neuroscience, School for Mental Health and Neuroscience, University of Maastricht, Maastricht, The Netherlands.
| | - Kenneth B Chapman
- Department of Anesthesiology, New York University Langone Medical Center, New York, NY, USA; Department of Anesthesiology, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Chris Terwiel
- Department of Anesthesiology and Pain Management Arnhem, Rijnstate Hospital, Arnhem, The Netherlands
| | - Elbert A Joosten
- Department of Anesthesiology and Pain Management, University Pain Clinic Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands; Department of Translational Neuroscience, School for Mental Health and Neuroscience, University of Maastricht, Maastricht, The Netherlands
| | - Jan Willem Kallewaard
- Department of Anesthesiology and Pain Management Arnhem, Rijnstate Hospital, Arnhem, The Netherlands; Department of Anesthesiology and Pain Management, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Sivanesan E, North RB, Russo MA, Levy RM, Linderoth B, Hayek SM, Eldabe S, Lempka SF. A Definition of Neuromodulation and Classification of Implantable Electrical Modulation for Chronic Pain. Neuromodulation 2024; 27:1-12. [PMID: 37952135 DOI: 10.1016/j.neurom.2023.10.004] [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: 08/16/2023] [Revised: 09/24/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023]
Abstract
OBJECTIVES Neuromodulation therapies use a variety of treatment modalities (eg, electrical stimulation) to treat chronic pain. These therapies have experienced rapid growth that has coincided with escalating confusion regarding the nomenclature surrounding these neuromodulation technologies. Furthermore, studies are often published without a complete description of the effective stimulation dose, making it impossible to replicate the findings. To improve clinical care and facilitate dissemination among the public, payors, research groups, and regulatory bodies, there is a clear need for a standardization of terms. APPROACH We formed an international group of authors comprising basic scientists, anesthesiologists, neurosurgeons, and engineers with expertise in neuromodulation. Because the field of neuromodulation is extensive, we chose to focus on creating a taxonomy and standardized definitions for implantable electrical modulation of chronic pain. RESULTS We first present a consensus definition of neuromodulation. We then describe a classification scheme based on the 1) intended use (the site of modulation and its indications) and 2) physical properties (waveforms and dose) of a neuromodulation therapy. CONCLUSIONS This framework will help guide future high-quality studies of implantable neuromodulatory treatments and improve reporting of their findings. Standardization with this classification scheme and clear definitions will help physicians, researchers, payors, and patients better understand the applications of implantable electrical modulation for pain and guide informed treatment decisions.
Collapse
Affiliation(s)
- Eellan Sivanesan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| | - Richard B North
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA; Department of Neurosurgery, Johns Hopkins University, Baltimore, MD, USA
| | - Marc A Russo
- Hunter Pain Specialists, Broadmeadow, New South Wales, Australia
| | - Robert M Levy
- Neurosurgical Services, Clinical Research, Anesthesia Pain Care Consultants, Tamarac, FL, USA
| | - Bengt Linderoth
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Salim M Hayek
- Division of Pain Medicine, University Hospitals, Cleveland Medical Center, Cleveland, OH, USA
| | - Sam Eldabe
- Department of Pain Medicine, The James Cook University Hospital, Middlesbrough, UK
| | - Scott F Lempka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
14
|
Lu L, Lau M, Akers L, Jones L, Selassie M, Burke M, Barley J, Hillegass M, Gleichgerrcht E. Intraoperative Neurophysiological Monitoring During Lead Placement for Dorsal Root Ganglion Stimulation: A Literature Review and Case Series. Neuromodulation 2024; 27:160-171. [PMID: 37245141 DOI: 10.1016/j.neurom.2023.04.468] [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: 12/22/2022] [Revised: 03/23/2023] [Accepted: 04/11/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Dorsal root ganglion stimulation (DRG-S) is a viable interventional option for intractable pain management. Although systematic data are lacking regarding the immediate neurologic complications of this procedure, intraoperative neurophysiological monitoring (IONM) can be a valuable tool to detect real-time neurologic changes and prompt intervention(s) during DRG-S performed under general anesthesia and deep sedation. MATERIALS AND METHODS In our single-center case series, we performed multimodal IONM, including peripheral nerve somatosensory evoked potentials (pnSSEPs) and dermatomal somatosensory evoked potentials (dSSEPs), spontaneous electromyography (EMG), transcranial motor evoked potentials (MEPs), and electroencephalogram (EEG) for some trials and all permanent DRG-S lead placement per surgeon preference. Alert criteria for each IONM modality were established before data acquisition and collection. An IONM alert was used to implement an immediate repositioning of the lead to reduce any possible postoperative neurologic deficits. We reviewed the literature and summarized the current IONM modalities commonly applied during DRG-S, including somatosensory evoked potentials and EMG. Because DRG-S targets the dorsal roots, we hypothesized that including dSSEP would allow more sensitivity as a proxy for potential sensory changes under generalized anesthesia than would including standard pnSSEPs. RESULTS From our case series of 22 consecutive procedures with 45 lead placements, one case had an alert immediately after DRG-S lead positioning. In this case, dSSEP attenuation was seen, indicating changes in the S1 dermatome, which occurred despite ipsilateral pnSSEP from the posterior tibial nerve remaining at baselines. The dSSEP alert prompted the surgeon to reposition the S1 lead, resulting in immediate recovery of the dSSEP to baseline status. The rate of IONM alerts reported intraoperatively was 4.55% per procedure and 2.22% per lead (n = 1). No neurologic deficits were reported after the procedure, resulting in no postoperative neurologic complications or deficits. No other IONM changes or alerts were observed from pnSSEP, spontaneous EMG, MEPs, or EEG modalities. Reviewing the literature, we noted challenges and potential deficiencies when using current IONM modalities for DRG-S procedures. CONCLUSIONS Our case series suggests dSSEPs offer greater reliability than do pnSSEPs in quickly detecting neurologic changes, and subsequent neural injury, during DRG-S cases. We encourage future studies to focus on adding dSSEP to standard pnSSEP to provide a comprehensive, real-time neurophysiological assessment during lead placement for DRG-S. More investigation, collaboration, and evidence are required to evaluate, compare, and standardize comprehensive IONM protocols for DRG-S.
Collapse
Affiliation(s)
- Lei Lu
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA.
| | - Martha Lau
- Department of Clinical Neurophysiology, Medical University of South Carolina, Charleston, SC, USA
| | - Lindsey Akers
- Department of Clinical Neurophysiology, Medical University of South Carolina, Charleston, SC, USA
| | - Lakota Jones
- Department of Clinical Neurophysiology, Medical University of South Carolina, Charleston, SC, USA
| | - Meron Selassie
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Martin Burke
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Jessica Barley
- Department of Clinical Neurophysiology, Medical University of South Carolina, Charleston, SC, USA
| | - Michael Hillegass
- Department of Anesthesiology and Perioperative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
15
|
Mol F, Scheltinga M, Roumen R, Wille F, Gültuna I, Kallewaard JW, Elzinga L, van de Minkelis J, Nijhuis H, Stronks DL, Huygen FJPM. Comparing the Efficacy of Dorsal Root Ganglion Stimulation With Conventional Medical Management in Patients With Chronic Postsurgical Inguinal Pain: Post Hoc Analyzed Results of the SMASHING Study. Neuromodulation 2023; 26:1788-1794. [PMID: 36456417 DOI: 10.1016/j.neurom.2022.09.014] [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: 08/25/2022] [Accepted: 09/28/2022] [Indexed: 12/03/2022]
Abstract
OBJECTIVES Approximately 10% of patients who undergo inguinal hernia repair or Pfannenstiel incision develop chronic (> three months) postsurgical inguinal pain (PSIP). If medication or peripheral nerve blocks fail, a neurectomy is the treatment of choice. However, some patients do not respond to this treatment. In such cases, stimulation of the dorsal root ganglion (DRG) appears to significantly reduce chronic PSIP in selected patients. MATERIALS AND METHODS In this multicenter, randomized controlled study, DRG stimulation was compared with conventional medical management (CMM) (noninvasive treatments, such as medication, transcutaneous electric neurostimulation, and rehabilitation therapy) in patients with PSIP that was resistant to a neurectomy. Patients were recruited at a tertiary referral center for groin pain (SolviMáx, Eindhoven, The Netherlands) between March 2015 and November 2016. Suitability for implantation was assessed according to the Dutch Neuromodulation Association guidelines. The sponsor discontinued the study early owing to slow enrollment. Of 78 planned patients, 18 were randomized (DRG and CMM groups each had nine patients). Six patients with CMM (67%) crossed over to DRG stimulation at the six-month mark. RESULTS Fifteen of the 18 patients met the six-month primary end point with a complete data set for a per-protocol analysis. Three patients with DRG stimulation had a negative trial and were lost to follow-up. The average pain reduction was 50% in the DRG stimulation and crossover group (from 6.60 ± 1.24 to 3.28 ± 2.30, p = 0.0029). Conversely, a 13% increase in pain was observed in patients with CMM (from 6.13 ± 2.24 to 6.89 ± 1.24, p = 0.42). Nine patients with DRG stimulation experienced a total of 19 adverse events, such as lead dislocation and pain at the implantation site. CONCLUSIONS DRG stimulation is a promising effective therapy for pain relief in patients with PSIP resistant to conventional treatment modalities; larger studies should confirm this. The frequency of side effects should be a concern in a new study. CLINICAL TRIAL REGISTRATION The Clinicaltrials.gov registration number for the study is NCT02349659.
Collapse
Affiliation(s)
| | | | - Rudi Roumen
- Maxima Medical Center, Eindhoven, The Netherlands
| | - Frank Wille
- Maxima Medical Center, Eindhoven, The Netherlands
| | | | | | - Lars Elzinga
- Maxima Medical Center, Eindhoven, The Netherlands
| | | | | | | | | |
Collapse
|
16
|
Arciero E, Coury JR, Dionne A, Reyes J, Lombardi JM, Sardar ZM. Optimizing Preoperative Chronic Pain Management in Elective Spine Surgery Patients: A Narrative Review of Outcomes with Opioid and Adjuvant Pain Therapies. JBJS Rev 2023; 11:01874474-202312000-00006. [PMID: 38100612 DOI: 10.2106/jbjs.rvw.23.00156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
» Chronic preoperative opioid use negatively affects outcomes after spine surgery, with increased complications and reoperations, longer hospital stays, decreased return-to-work rates, worse patient-reported outcomes, and a higher risk of continued opioid use postoperatively.» The definition of chronic opioid use is not consistent across studies, and a more specific and consistent definition will aid in stratifying patients and understanding their risk of inferior outcomes.» Preoperative weaning periods and maximum dose thresholds are being established, which may increase the likelihood of achieving a meaningful improvement after surgery, although higher level evidence studies are needed.» Spinal cord stimulators and intrathecal drug delivery devices are increasingly used to manage chronic back pain and are equivalent or perhaps even superior to opioid treatment, although few studies exist examining how patients with these devices do after subsequent spine surgery.» Further investigation is needed to determine whether a true mechanistic explanation exists for spine-related analgesia related to spinal cord stimulators and intrathecal drug delivery devices.
Collapse
Affiliation(s)
- Emily Arciero
- The Och Spine Hospital, New York-Presbyterian/Columbia University Irving Medical Center, New York, New York
| | | | | | | | | | | |
Collapse
|
17
|
Cho YH, Seo TB. The role of walking exercise on axonal regrowth and neuropathic pain markers in dorsal root ganglion after sciatic nerve injury. J Exerc Rehabil 2023; 19:320-326. [PMID: 38188130 PMCID: PMC10766449 DOI: 10.12965/jer.2346522.261] [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: 10/06/2023] [Accepted: 11/13/2023] [Indexed: 01/09/2024] Open
Abstract
The aim of this study was to determine whether walking exercise can regulate the expression level of neuropathic pain- and inflammatory response markers in the ipsilateral lumbar 4 to 6 dorsal root ganglion neurons after sciatic nerve injury (SNI). The experimental rats were randomly divided into seven groups: the normal control group, sedentary groups for 3, 7, and, 14 days postinjury (dpi), and walking exercise groups for 3, 7, and 14 dpi. Western blot techniques were used to evaluate specific neuropathic pain- and cytokine markers and mechanical allodynia was confirmed by paw withdrawal test. Mechanical allodynia was significantly improved in the walking exercise group compared to the sedentary group at all 7, 10, and 14 dpi. Furthermore, growth associated protein 43 and brain-derived neurotrophic factor levels were significantly increased in the walking exercise groups compared to the sedentary group at all 3, 7, and 14 dpi. Conversely, nuclear factor kappa-light-chain-enhancer of activated B cells, interleukin-6, tumor necrosis factor α, calcitonin gene-related peptide, and c-Fos expression levels were significantly decreased in the walking exercise groups compared to the sedentary group at all 3, 7, and 14 dpi. These findings suggest meaningful information that aggressive rehabilitation walking exercise applied early after SNI might be improve mechanical allodynia, neuropathic pain and inflammatory response markers following SNI.
Collapse
Affiliation(s)
- Yeong-Hyun Cho
- Department of Kinesiology, College of Natural Science, Jeju National University, Jeju,
Korea
| | - Tae-Beom Seo
- Department of Kinesiology, College of Natural Science, Jeju National University, Jeju,
Korea
| |
Collapse
|
18
|
Koetsier E, Vacchi E, Maino P, Dukanac J, Melli G, van Kuijk SMJ. Dorsal Root Ganglion Stimulation in Chronic Painful Polyneuropathy: A Potential Modulator for Small Nerve Fiber Regeneration. Neuromodulation 2023; 26:1772-1780. [PMID: 36192280 DOI: 10.1016/j.neurom.2022.08.455] [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: 05/09/2022] [Revised: 08/02/2022] [Accepted: 08/23/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVES Neuromodulatory treatments like spinal cord stimulation and dorsal root ganglion stimulation (DRGS) have emerged as effective treatments to relieve pain in painful polyneuropathy. Animal studies have demonstrated that neurostimulation can enhance nerve regeneration. This study aimed to investigate if DRGS may impact intraepidermal nerve fiber regeneration and sensory nerve function. MATERIALS AND METHODS Nine patients with chronic, intractable painful polyneuropathy were recruited. Intraepidermal nerve fiber density (IENFD) quantification in 3 mm punch skin biopsy was performed 1 month before DRGS (placed at the level of the L5 and S1 dorsal root ganglion) and after 12- and 24-month follow-up. Quantitative sensory testing, nerve conduction studies, and a clinical scale score were also performed at the same time points. RESULTS In 7 of 9 patients, DRGS was successful (defined as a reduction of ≥ 50% in daytime and/or night-time pain intensity), allowing a definitive implantable pulse generator implantation. The median baseline IENFD among these 7 patients was 1.6 fibers/mm (first and third quartile: 1.2; 4.3) and increased to 2.6 fibers/mm (2.5; 2.9) and 1.9 fibers/mm (1.6; 2.4) at 1- and 2-years follow-up, respectively. These changes were not statistically significant (p = 1.000 and 0.375). Sensory nerve tests did not show substantial changes. CONCLUSIONS Although not significant, the results of this study showed that in most of the patients with implants, there was a slight increase of the IENFD at the 1- and 2-year follow-up. Larger-scale clinical trials are warranted to explore the possible role of DRGS in reversing the progressive neurodegeneration over time. CLINICAL TRIAL REGISTRATION The Clinicaltrials.gov registration number for the study is NCT02435004; Swiss National Clinical Trials Portal: SNCTP000001376.
Collapse
Affiliation(s)
- Eva Koetsier
- Pain Management Center, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland.
| | - Elena Vacchi
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Laboratories for Translational Research, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Paolo Maino
- Pain Management Center, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Jasmina Dukanac
- Pain Management Center, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Giorgia Melli
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Laboratories for Translational Research, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Neurology Department, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Sander M J van Kuijk
- Pain Management Center, Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland; Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
19
|
Homs M, Milà R, Valdés R, Blay D, Borràs RM, Parés D. Efficacy of conditioned autologous serum therapy (Orthokine®) on the dorsal root ganglion in patients with chronic radiculalgia: study protocol for a prospective randomized placebo-controlled double-blind clinical trial (RADISAC trial). Trials 2023; 24:755. [PMID: 38007491 PMCID: PMC10676602 DOI: 10.1186/s13063-023-07787-y] [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: 04/24/2023] [Accepted: 11/06/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Pulsed radiofrequency (PRF) treatment on the dorsal root ganglion (DRG) has been proposed as a good option for the treatment of persistent radicular pain based on its effect of neuromodulation on neuropathic pain. Autologous conditioned serum (ACS) therapy is a conservative treatment based on the patient's own blood. The aim of this manuscript is to develop a study protocol using ACS on the DRG as a target for its molecular modulation. METHODS We plan to conduct a randomized controlled study to compare the efficacy of PRF therapy plus ACS versus PRF therapy plus physiological saline 0.9% (PhS) on the DRG to reduce neuropathic pain in patients with persistent lower limb radiculalgia (LLR) and to contribute to the functional improvement and quality of life of these patients. Study participants will include patients who meet study the inclusion/exclusion criteria. Eligible patients will be randomized in a 1:1 ratio to one of treatment with PRF plus ACS (experimental group) or PRF plus PhS (placebo group). The study group will consist of 70 patients (35 per group) who have experienced radicular pain symptoms for ≥ 6 months' duration who have failed to respond to any therapy. Both groups will receive PRF on the DRG treatment before the injection of the sample (control or placebo). Patient assessments will occur at baseline, 1 month, 3 months, 6 months, and 12 months after therapy. The primary efficacy outcome measure is Numeric Pain Rating Scale (NPRS) responders from baseline to 12 months of follow-up using validated minimal important change (MIC) thresholds. A reduction of ≥ 2 points in NPRS is considered a clinically significant pain relief. The secondary efficacy outcome measure is the proportion of Oswestry Low Back Pain Disability Scale (ODS) responders from baseline to 12 months of follow-up in the experimental group (PRF plus ACS) versus the placebo group (PRF plus PhS). ODS responders are defined as those patients achieving the validated MIC of ≥ 10-point improvement in ODS from baseline to 12 months of follow-up as a clinically significant efficacy threshold. DISCUSSION This prospective, double-blind, randomized placebo-controlled study will provide level I evidence of the safety and effectiveness of ACS on neuropathic symptoms in LLR patients. TRIAL REGISTRATION {2A}{2B}: EUDRACT number: 2021-005124-38. Validation date: 13 November 2021. Protocol version {3}: This manuscript presents the 2nd protocol version.
Collapse
Affiliation(s)
- Marta Homs
- Dexeus University Hospital, Sabino Arana 5-19, 08028, Barcelona, Spain.
| | - Raimon Milà
- Ramon Llull University, Pg St Gerbasi 43, 08022, Barcelona, Spain
| | - Ricard Valdés
- Dexeus University Hospital, Sabino Arana 5-19, 08028, Barcelona, Spain
| | | | - Rosa Maria Borràs
- Dexeus University Hospital, Sabino Arana 5-19, 08028, Barcelona, Spain
| | - David Parés
- Hospital Germans Trias I Pujol, Carretera del Canyet S/N, 08916, Badalona, Spain
| |
Collapse
|
20
|
Berta T, Strong JA, Zhang JM, Ji RR. Targeting dorsal root ganglia and primary sensory neurons for the treatment of chronic pain: an update. Expert Opin Ther Targets 2023; 27:665-678. [PMID: 37574713 PMCID: PMC10530032 DOI: 10.1080/14728222.2023.2247563] [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: 04/04/2023] [Revised: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 08/15/2023]
Abstract
INTRODUCTION Current treatments for chronic pain are inadequate. Here, we provide an update on the new therapeutic strategies that target dorsal root ganglia (DRGs) in the peripheral nervous system for a better and safer treatment of chronic pain. AREAS COVERED Despite the complex nature of chronic pain and its underlying mechanisms, we do know that changes in the plasticity and modality of neurons in DRGs play a pivotal role. DRG neurons are heterogenous and offer potential pain targets for different therapeutic interventions. We discuss the last advancements of these interventions, which include the use of systemic and local administrations, selective nerve drug delivery, and gene therapy. In particular, we provide updates and further details on the molecular characterization of primary sensory neurons, new analgesics entering the market, and future gene therapy approaches. EXPERT OPINION DRGs and primary sensory neurons are promising targets for chronic pain treatment due to their key role in pain signaling, unique anatomical location, and the potential for different targeted therapeutic interventions.
Collapse
Affiliation(s)
- Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Judith A. Strong
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Jun-Ming Zhang
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH 45267, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
- Departments of Cell Biology and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
21
|
Burkey AR, Chen J, Argoff CE, Edgar DR, Petersen EA. Painful Peripheral Neuropathies of the Lower Limbs and/or Lower Extremities Treated with Spinal Cord Stimulation: A Systematic Review with Narrative Synthesis. J Pain Res 2023; 16:1607-1636. [PMID: 37229154 PMCID: PMC10202826 DOI: 10.2147/jpr.s403715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/21/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Painful peripheral neuropathy (PPN) is a debilitating condition with varied etiologies. Spinal cord stimulation (SCS) is increasingly used when conservative treatments fail to provide adequate pain relief. Few published reviews have examined SCS outcomes in all forms of PPN. Methods We conducted a systematic review of SCS in PPN. The PubMed database was searched up to February 7th, 2022, for peer-reviewed studies of SCS that enrolled PPN patients with pain symptoms in their lower limbs and/or lower extremities. We assessed the quality of randomized controlled trial (RCT) evidence using the Cochrane risk of bias tool. Data were tabulated and presented narratively. Results Twenty eligible studies documented SCS treatment in PPN patients, including 10 kHz SCS, traditional low-frequency SCS (t-SCS), dorsal root ganglion stimulation (DRGS), and burst SCS. In total, 451 patients received a permanent implant (10 kHz SCS, n=267; t-SCS, n=147; DRGS, n=25; burst SCS, n=12). Approximately 88% of implanted patients had painful diabetic neuropathy (PDN). Overall, we found clinically meaningful pain relief (≥30%) with all SCS modalities. Among the studies, RCTs supported the use of 10 kHz SCS and t-SCS to treat PDN, with 10 kHz SCS providing a higher reduction in pain (76%) than t-SCS (38-55%). Pain relief with 10 kHz SCS and DRGS in other PPN etiologies ranged from 42-81%. In addition, 66-71% of PDN patients and 38% of nondiabetic PPN patients experienced neurological improvement with 10 kHz SCS. Conclusion Our review found clinically meaningful pain relief in PPN patients after SCS treatment. RCT evidence supported the use of 10 kHz SCS and t-SCS in the diabetic neuropathy subpopulation, with more robust pain relief evident with 10 kHz SCS. Outcomes in other PPN etiologies were also promising for 10 kHz SCS. In addition, a majority of PDN patients experienced neurological improvement with 10 kHz SCS, as did a notable subset of nondiabetic PPN patients.
Collapse
Affiliation(s)
| | - Jeffrey Chen
- UCSD Department of Anesthesiology Center for Pain, University of California San Diego Medical Center, La Jolla, CA, USA
| | | | | | - Erika A Petersen
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
22
|
van Heteren EPZ, van Roosendaal BKWP, van Gorp EJJAA, Bronkhorst EM, Kallewaard JW, Wegener JT, Bürger K, Teernstra OPM, Buschman HPJ, Hamm-Faber TE, Vissers KCP. Spinal Cord Stimulation With Additional Peripheral Nerve/Field Stimulation Versus Spinal Cord Stimulation Alone on Back Pain and Quality of Life in Patients With Persistent Spinal Pain Syndrome. Neuromodulation 2023; 26:658-665. [PMID: 35088732 DOI: 10.1016/j.neurom.2021.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/04/2021] [Accepted: 10/31/2021] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Persistent spinal pain syndrome (PSPS) or failed back surgery syndrome (FBSS) refers to new or persistent pain following spinal surgery for back or leg pain in a subset of patients. Spinal cord stimulation (SCS) is a neuromodulation technique that can be considered in patients with predominant leg pain refractory to conservative treatment. Patients with predominant low back pain benefit less from SCS. Another neuromodulation technique for treatment of chronic low back pain is subcutaneous stimulation or peripheral nerve field stimulation (PNFS). We investigated the effect of SCS with additional PNFS on pain and quality of life of patients with PSPS compared with that of SCS alone after 12 months. MATERIALS AND METHODS This is a comparative study of patients with PSPS who responded to treatment with either SCS + PNFS or SCS only following a multicenter randomized clinical trial protocol. In total, 75 patients completed the 12-month follow-up: 21 in the SCS-only group and 54 in the SCS + PNFS group. Outcome measures were pain (visual analog scale), quality of life (36-Item Short Form Survey [SF-36]), anxiety and depression (Hospital Anxiety and Depression Scale [HADS]), overall health (EuroQol Five-Dimension [EQ-5D]), disability (Oswestry Disability Index [ODI]), and pain assessed by the McGill questionnaire. RESULTS There were no significant differences in baseline characteristics between the two groups. Both groups showed a significant reduction in back and leg pain at 12 months compared with baseline measurements. No significant differences were found between the groups in effect on both primary (pain) and secondary parameters (SF-36, HADS, EQ-5D, ODI, and McGill pain). CONCLUSION In a subgroup of patients with chronic back and leg pain, SCS alone provided similar long-term pain relief and quality-of-life improvement as PNFS in addition to SCS. In patients with refractory low back pain not responding to SCS alone, adding PNFS should be recommended. CLINICAL TRIAL REGISTRATION The Clinicaltrials.gov registration number for the study is NCT01776749.
Collapse
Affiliation(s)
- Esther P Z van Heteren
- Department of Anesthesiology, Pain, and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | | | - Eric-Jan J A A van Gorp
- Department of Anesthesiology, Unit of Pain Medicine, Albert Schweitzer Hospital, Sliedrecht, The Netherlands
| | - Ewald M Bronkhorst
- Department of Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Jessica T Wegener
- Department of Anesthesiology and Pain Medicine, Sint Maartenskliniek, Nijmegen, The Netherlands
| | - Katja Bürger
- Department of Anesthesiology, Rijnland Hospital, Leiderdorp, The Netherlands
| | - Otto P M Teernstra
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | - Tanja E Hamm-Faber
- Department of Pain Medicine, Albert Schweitzer Hospital, Zwijndrecht, The Netherlands
| | - Kris C P Vissers
- Department of Anesthesiology, Pain, and Palliative Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
23
|
Chapman KB, Sayed D, Lamer T, Hunter C, Weisbein J, Patel KV, Dickerson D, Hagedorn JM, Lee DW, Amirdelfan K, Deer T, Chakravarthy K. Best Practices for Dorsal Root Ganglion Stimulation for Chronic Pain: Guidelines from the American Society of Pain and Neuroscience. J Pain Res 2023; 16:839-879. [PMID: 36942306 PMCID: PMC10024474 DOI: 10.2147/jpr.s364370] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 01/17/2023] [Indexed: 03/14/2023] Open
Abstract
With continued innovations in neuromodulation comes the need for evolving reviews of best practices. Dorsal root ganglion stimulation (DRG-S) has significantly improved the treatment of complex regional pain syndrome (CRPS), and it has broad applicability across a wide range of other conditions. Through funding and organizational leadership by the American Society for Pain and Neuroscience (ASPN), this best practices consensus document has been developed for the selection, implantation, and use of DRG stimulation for the treatment of chronic pain syndromes. This document is composed of a comprehensive narrative literature review that has been performed regarding the role of the DRG in chronic pain and the clinical evidence for DRG-S as a treatment for multiple pain etiologies. Best practice recommendations encompass safety management, implantation techniques, and mitigation of the potential complications reported in the literature. Looking to the future of neuromodulation, DRG-S holds promise as a robust intervention for otherwise intractable pain.
Collapse
Affiliation(s)
- Kenneth B Chapman
- The Spine & Pain Institute of New York, New York, NY, USA
- Department of Anesthesiology, Zucker School of Medicine at Hofstra Northwell, Manhasset, NY, USA
- Department of Anesthesiology, NYU Langone Medical Center, New York, NY, USA
| | - Dawood Sayed
- Department of Anesthesiology, The University of Kansas Medical Center (KUMC), Kansas City, KS, USA
| | - Tim Lamer
- Department of Anesthesiology and Perioperative Medicine, Division of Pain Medicine, Mayo Clinic, Rochester, MN, USA
| | - Corey Hunter
- Ainsworth Institute of Pain Management, New York, NY, USA
| | | | - Kiran V Patel
- The Spine & Pain Institute of New York, New York, NY, USA
- Department of Anesthesiology, Zucker School of Medicine at Hofstra Northwell, Manhasset, NY, USA
- Department of Anesthesiology, NYU Langone Medical Center, New York, NY, USA
| | - David Dickerson
- Department of Anesthesiology, Critical Care and Pain Medicine, NorthShore University Health System, Evanston, IL, USA
- Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL, USA
| | | | - David W Lee
- Fullerton Orthopedic Surgery Medical Group, Fullerton, CA, USA
| | | | - Timothy Deer
- The Spine and Nerve Center of the Virginias, Charleston, WV, USA
| | - Krishnan Chakravarthy
- Department of Anesthesiology and Pain Medicine, University of California San Diego Health Sciences, San Diego, CA, USA
- VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
24
|
Medina R, Ho A, Reddy R, Chen J, Castellanos J. Narrative review of current neuromodulation modalities for spinal cord injury. FRONTIERS IN PAIN RESEARCH 2023; 4:1143405. [PMID: 36969918 PMCID: PMC10033643 DOI: 10.3389/fpain.2023.1143405] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
Neuromodulation is a developing field of medicine that includes a vast array of minimally invasive and non-invasive therapies including transcranial magnetic stimulation (TMS), transcranial direct current stimulation (tDCS), vagus nerve stimulation (VNS), peripheral nerve stimulation, and spinal cord stimulation (SCS). Although the current literature surrounding the use of neuromodulation in managing chronic pain is abundant, there is an insufficient amount of evidence specifically regarding neuromodulation in patients with spinal cord injury (SCI). Given the pain and functional deficits that these patients face, that are not amenable to other forms conservative therapy, the purpose of this narrative review is to examine and assess the use of various neuromodulation modalities to manage pain and restore function in the SCI population. Currently, high-frequency spinal cord stimulation (HF-SCS) and burst spinal cord stimulation (B-SCS) have been shown to have the most promising effect in improving pain intensity and frequency. Additionally, dorsal root ganglion stimulation (DRG-S) and TMS have been shown to effectively increase motor responses and improve limb strength. Although these modalities carry the potential to enhance overall functionality and improve a patient's degree of disability, there is a lack of long-term, randomized-controlled trials in the current space. Additional research is warranted to further support the clinical use of these emerging modalities to provide improved pain management, increased level of function, and ultimately an overall better quality of life in the SCI population.
Collapse
Affiliation(s)
- Roi Medina
- Department of Physical Medicine and Rehabilitation, Lake Erie College of Osteopathic Medicine-Bradenton, Bradenton, FL, United States
| | - Alison Ho
- Department of Physical Medicine and Rehabilitation, Baylor University Medical Center, Dallas, TX, United States
| | - Rajiv Reddy
- UC San Diego Health, University of California San Diego, La Jolla, CA, United States
| | - Jeffrey Chen
- UC San Diego Health, University of California San Diego, La Jolla, CA, United States
| | - Joel Castellanos
- UC San Diego Health, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
25
|
Hao H, Ramli R, Wang C, Liu C, Shah S, Mullen P, Lall V, Jones F, Shao J, Zhang H, Jaffe DB, Gamper N, Du X. Dorsal root ganglia control nociceptive input to the central nervous system. PLoS Biol 2023; 21:e3001958. [PMID: 36603052 PMCID: PMC9847955 DOI: 10.1371/journal.pbio.3001958] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 01/18/2023] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Accumulating observations suggest that peripheral somatosensory ganglia may regulate nociceptive transmission, yet direct evidence is sparse. Here, in experiments on rats and mice, we show that the peripheral afferent nociceptive information undergoes dynamic filtering within the dorsal root ganglion (DRG) and suggest that this filtering occurs at the axonal bifurcations (t-junctions). Using synchronous in vivo electrophysiological recordings from the peripheral and central processes of sensory neurons (in the spinal nerve and dorsal root), ganglionic transplantation of GABAergic progenitor cells, and optogenetics, we demonstrate existence of tonic and dynamic filtering of action potentials traveling through the DRG. Filtering induced by focal application of GABA or optogenetic GABA release from the DRG-transplanted GABAergic progenitor cells was specific to nociceptive fibers. Light-sheet imaging and computer modeling demonstrated that, compared to other somatosensory fiber types, nociceptors have shorter stem axons, making somatic control over t-junctional filtering more efficient. Optogenetically induced GABA release within DRG from the transplanted GABAergic cells enhanced filtering and alleviated hypersensitivity to noxious stimulation produced by chronic inflammation and neuropathic injury in vivo. These findings support "gating" of pain information by DRGs and suggest new therapeutic approaches for pain relief.
Collapse
Affiliation(s)
- Han Hao
- Department of Pharmacology, Hebei Medical University; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Rosmaliza Ramli
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- School of Dental Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Caixue Wang
- Department of Pharmacology, Hebei Medical University; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Chao Liu
- Department of Animal Care, Hebei Medical University; The Key Laboratory of Experimental Animal, Hebei Province; Shijiazhuang, China
| | - Shihab Shah
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Pierce Mullen
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Varinder Lall
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Frederick Jones
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Jicheng Shao
- Department of Pharmacology, Hebei Medical University; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - David B. Jaffe
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| |
Collapse
|
26
|
Pricope CV, Tamba BI, Stanciu GD, Cuciureanu M, Neagu AN, Creanga-Murariu I, Dobrovat BI, Uritu CM, Filipiuc SI, Pricope BM, Alexa-Stratulat T. The Roles of Imaging Biomarkers in the Management of Chronic Neuropathic Pain. Int J Mol Sci 2022; 23:13038. [PMID: 36361821 PMCID: PMC9657736 DOI: 10.3390/ijms232113038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/22/2022] [Accepted: 10/24/2022] [Indexed: 08/04/2023] Open
Abstract
Chronic neuropathic pain (CNP) affects around 10% of the general population and has a significant social, emotional, and economic impact. Current diagnosis techniques rely mainly on patient-reported outcomes and symptoms, which leads to significant diagnostic heterogeneity and subsequent challenges in management and assessment of outcomes. As such, it is necessary to review the approach to a pathology that occurs so frequently, with such burdensome and complex implications. Recent research has shown that imaging methods can detect subtle neuroplastic changes in the central and peripheral nervous system, which can be correlated with neuropathic symptoms and may serve as potential markers. The aim of this paper is to review available imaging methods used for diagnosing and assessing therapeutic efficacy in CNP for both the preclinical and clinical setting. Of course, further research is required to standardize and improve detection accuracy, but available data indicate that imaging is a valuable tool that can impact the management of CNP.
Collapse
Affiliation(s)
- Cosmin Vasilica Pricope
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Department of Pharmacology, Clinical Pharmacology and Algesiology, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Bogdan Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Department of Pharmacology, Clinical Pharmacology and Algesiology, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Gabriela Dumitrita Stanciu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Magdalena Cuciureanu
- Department of Pharmacology, Clinical Pharmacology and Algesiology, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Anca Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, Carol I bvd. No. 22, 700505 Iasi, Romania
| | - Ioana Creanga-Murariu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Bogdan-Ionut Dobrovat
- Department of Radiology, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 University Street, 700115 Iasi, Romania
| | - Cristina Mariana Uritu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Silviu Iulian Filipiuc
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Bianca-Mariana Pricope
- Department of Preventive Medicine and Interdisciplinarity, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
| | - Teodora Alexa-Stratulat
- Advanced Research and Development Center for Experimental Medicine (CEMEX), Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115 Iasi, Romania
- Medical Oncology-Radiotherapy Department, Grigore T. Popa University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| |
Collapse
|
27
|
Telkes I, Hadanny A, DiMarzio M, Chitnis G, Paniccioli S, O'Connor K, Grey R, McCarthy K, Khazen O, McLaughlin B, Pilitsis JG. High-Resolution Spinal Motor Mapping Using Thoracic Spinal Cord Stimulation in Patients With Chronic Pain. Neurosurgery 2022; 91:459-469. [PMID: 35876669 PMCID: PMC10553191 DOI: 10.1227/neu.0000000000002054] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 04/11/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND High-resolution spinal cord stimulation (HR-SCS) paddle can stimulate medial-dorsal columns and extend stimulation coverage to the laterally positioned spinal targets. OBJECTIVE To investigate the medio-lateral selectivity of an HR-SCS paddle in patients with chronic pain. METHODS During standard-of-care spinal cord stimulation (SCS) placement, epidurally evoked electromyography and antidromic dorsal column-evoked potentials were recorded in 12 subjects using an HR-SCS paddle with 8 medio-lateral sites spanning the full epidural width at thoracic T9-12 and a commercial paddle consecutively. RESULTS Recruitment maps were aligned with respect to physiological midline which was overlapping with anatomic midline in 10 of 11 cases. Overlapping contacts between the HR-SCS and commercial paddles exhibited similar patterns while HR-SCS demonstrated higher precision targeting of certain dermatomes. Spinal motor maps showed that the lateral contacts triggered stronger responses in medial gastrocnemius, adductor magnus, and tibialis anterior while the medial contacts triggered stronger responses in gluteus maximus and adductor hallucis. The time-locked popliteal fossa responses indicated ipsilateral activation by HR-SCS at the lateral contacts and bilateral activation at the medial contacts with stronger ipsilateral responses. CONCLUSION This study is the first to perform high-resolution medio-lateral SCS mapping in patients with chronic pain. These results show promise that HR-SCS may provide additional ipsilateral recruitment within the extremities which improve targeting of focal pain in the lower extremities. Furthermore, this study supports the functional use of intraoperative neuromonitoring as a decision tool to determine physiological midline in thoracic SCS surgeries and provides a full methodological framework.
Collapse
Affiliation(s)
- Ilknur Telkes
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Amir Hadanny
- Department of Neurosurgery, Albany Medical College, Albany, New York, USA
| | - Marisa DiMarzio
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | | | | | | | - Rachael Grey
- Nuvasive Clinical Services, San Diego, California, USA
| | | | - Olga Khazen
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | | | - Julie G. Pilitsis
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
- Department of Neurosurgery, Albany Medical College, Albany, New York, USA
| |
Collapse
|
28
|
|
29
|
Potter ST, Welch S, Tata F, Probert S, Nagpal A. Dorsal Root Ganglion Stimulation. Phys Med Rehabil Clin N Am 2022; 33:359-378. [DOI: 10.1016/j.pmr.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
30
|
Sheldon BL, Bao J, Khazen O, Pilitsis JG. Spinal Cord Stimulation as Treatment for Cancer and Chemotherapy-Induced Pain. FRONTIERS IN PAIN RESEARCH 2022; 2:699993. [PMID: 35295456 PMCID: PMC8915692 DOI: 10.3389/fpain.2021.699993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 07/23/2021] [Indexed: 01/17/2023] Open
Abstract
Neuropathic pain is a rampant disease exacting a significant toll on patients, providers, and health care systems around the globe. Neuromodulation has been successfully employed to treat many indications including failed back surgery syndrome (FBSS), complex regional pain syndrome (CRPS), phantom limb pain (PLP), radiculopathies, and intractable pelvic pain, among many others. Recent studies have also demonstrated efficacy for cancer-related pain and chemotherapy induced neuropathy with these techniques. Spinal cord stimulation (SCS) is the most commonly employed technique and involves implantation of percutaneous or paddle leads targeting the dorsal columns of the spinal cord with the goal of disrupting the pain signals traveling to the brain. Tonic, high frequency, and burst waveforms have all been shown to reduce pain and disability in chronic pain patients. Closed-loop SCS systems that automatically adjust stimulation parameters based on feedback (such as evoked compound action potentials) are becoming increasingly used to help ease the burden placed on patients to adjust their programming to their pain and position. Additionally, dorsal root ganglion stimulation (DRGS) is a newer technique that allows for dermatomal coverage especially in patients with pain in up to two dermatomes. Regardless of the technique chosen, neuromodulation has been shown to be cost-effective and efficacious and should be given full consideration in patients with chronic pain conditions.
Collapse
Affiliation(s)
- Breanna L Sheldon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Jonathan Bao
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Olga Khazen
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Julie G Pilitsis
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States.,Department of Neurosurgery, Albany Medical Center, Albany, NY, United States
| |
Collapse
|
31
|
Onwudiwe N, Charter R, Gingles B, Abrishami P, Alder H, Bahkai A, Civic D, Kosaner Kliess M, Lessard C, Zema C. Generating Appropriate and Reliable Evidence for Value Assessment of Medical Devices: An Ispor Medical Devices and Diagnostics Special Interest Group Report. J Med Device 2022. [DOI: 10.1115/1.4053928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Abstract
Background: Health Technology Assessment methods have become an important health policy tool. Yet recommendations for what constitutes appropriate and reliable evidence for assessment of medical devices are still debated because methods to evaluate pharmaceuticals are often, and incorrectly, the starting point for assessments.
Objectives:
The study aims to: (i) propose recommendations on appropriate methodologies to assess the evidence on medical devices (ii) identify assessment methods that can be used to measure device value and (iii) suggest key areas for future work
Methods:
ISPOR's Medical Devices and Diagnostics Special Interest Group conducted a comprehensive search of databases and gray literature on evidence development and value assessment on medical devices. The literature search was supplemented with hand searching from high impact journals in the related field. The 10-person expert working group obtained written comments through multiple rounds of review from internal and external stakeholders. Recommendations were made to guide future research.
Results:
Multi-criteria decision analysis was identified as a useful approach to assess the value of treatment. Consideration should be given to resource use measures; valid and reliable functional status questionnaires; and general and disease-specific, health-related, quality-of-life measures in economic evaluations of device use. For future work, best practices for value framework design.
Conclusions:
Integration of value-based evidence in an evidence-generation and -synthesis process is needed to support market access and adoption. Methodological recommendations for measuring value can be challenging when the selection of domains and assessment of value are not device-specific.
Collapse
Affiliation(s)
- Nneka Onwudiwe
- Pharmaceutical Economics Consultants of America, Silver Spring, MD, USA
| | | | | | | | - Henry Alder
- Access to Care Partners, LLC, Chicago, IL, USA
| | | | | | | | | | | |
Collapse
|
32
|
Prospective Observational Cohort Study on Dorsal Root Ganglion Stimulation in Chronic Postsurgical Pain: Results of Patient-Reported Outcomes at Two Years. Neuromodulation 2022; 25:998-1005. [DOI: 10.1016/j.neurom.2021.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 11/20/2022]
|
33
|
Rigoard P, Roulaud M, Goudman L, Adjali N, Ounajim A, Voirin J, Perruchoud C, Bouche B, Page P, Guillevin R, Naudin M, Simoneau M, Lorgeoux B, Baron S, Nivole K, Many M, Maitre I, Rigoard R, David R, Moens M, Billot M. Comparison of Spinal Cord Stimulation vs. Dorsal Root Ganglion Stimulation vs. Association of Both in Patients with Refractory Chronic Back and/or Lower Limb Neuropathic Pain: An International, Prospective, Randomized, Double-Blinded, Crossover Trial (BOOST-DRG Study). MEDICINA (KAUNAS, LITHUANIA) 2021; 58:7. [PMID: 35056316 PMCID: PMC8780129 DOI: 10.3390/medicina58010007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/01/2021] [Accepted: 12/15/2021] [Indexed: 12/25/2022]
Abstract
While spinal cord stimulation (SCS) is a well-established therapy to address refractory persistent spinal pain syndrome after spinal surgery (PSPS-T2), its lack of spatial selectivity and reported discomfort due to positional effects can be considered as significant limitations. As alternatives, new waveforms, such as burst stimulation and different spatial neural targets, such as dorsal root ganglion stimulation (DRGS), have shown promising results. Comparisons between DRGS and standard SCS, or their combination, have never been studied on the same patients. "BOOST DRG" is the first prospective, randomized, double-blinded, crossover study to compare SCS vs. DRGS vs. SCS+DRGS. Sixty-six PSPS-T2 patients will be recruited internationally in three centers. Before crossing over, patients will receive each stimulation modality for 1 month, using tonic conventional stimulation. After 3 months, stimulation will consist in switching to burst for 1 month, and patients will choose which modality/waveform they receive and will then be reassessed at 6 and 12 months. In addition to our primary outcome based on pain rating, this study is designed to assess quality of life, functional disability, psychological distress, pain surface coverage, global impression of change, medication quantification, adverse events, brain functional imaging and electroencephalography, with the objective being to provide a multidimensional insight based on composite pain assessment.
Collapse
Affiliation(s)
- Philippe Rigoard
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
- Department of Spine Surgery & Neuromodulation, Poitiers University Hospital, 86021 Poitiers, France;
- Pprime Institute UPR 3346, CNRS, ISAE-ENSMA, University of Poitiers, 86360 Chasseneuil-du-Poitou, France
| | - Manuel Roulaud
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
| | - Lisa Goudman
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium; (L.G.); (M.M.)
- STUMULUS Research Group, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Nihel Adjali
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
| | - Amine Ounajim
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
| | - Jimmy Voirin
- Department of Neurosurgery, Hopitaux Civils de Colmar, 68000 Colmar, France;
| | - Christophe Perruchoud
- Service of Anesthesiology and Pain Centre, University Hospital of Lausanne (CHUV), 1011 Lausanne, Switzerland;
| | - Bénédicte Bouche
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
- Department of Spine Surgery & Neuromodulation, Poitiers University Hospital, 86021 Poitiers, France;
| | - Philippe Page
- Department of Spine Surgery & Neuromodulation, Poitiers University Hospital, 86021 Poitiers, France;
| | - Rémy Guillevin
- Department of Radiology, Poitiers University Hospital, 86021 Poitiers, France; (R.G.); (M.N.)
- UMR CNRS 7348, DACTIM-MIS/LMA Laboratory, University of Poitiers, 86000 Poitiers, France
| | - Mathieu Naudin
- Department of Radiology, Poitiers University Hospital, 86021 Poitiers, France; (R.G.); (M.N.)
- UMR CNRS 7348, DACTIM-MIS/LMA Laboratory, University of Poitiers, 86000 Poitiers, France
| | - Martin Simoneau
- Department of Kinesiology, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada;
- Centre Interdisciplinaire de Recherche en Réadaptation et Intégration Sociale (CIRRIS), Quebec, QC G1M 2S8, Canada
| | - Bertille Lorgeoux
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
| | - Sandrine Baron
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
| | - Kevin Nivole
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
| | - Mathilde Many
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
| | - Iona Maitre
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
| | - Raphaël Rigoard
- CEA Cadarache, Département de Support Technique et Gestion, Service des Technologies de l’Information et de la Communication, 13108 Saint-Paul-Lez-Durance, France;
| | - Romain David
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
- Department of Physical and Rehabilitation Medicine, Poitiers University Hospital, University of Poitiers, 86021 Poitiers, France
| | - Maarten Moens
- Department of Neurosurgery, Universitair Ziekenhuis Brussel, 1090 Brussels, Belgium; (L.G.); (M.M.)
- STUMULUS Research Group, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Maxime Billot
- PRISMATICS Lab (Predictive Research in Spine/Neuromodulation Management and Thoracic Innovation/Cardiac Surgery), Poitiers University Hospital, 86021 Poitiers, France; (M.R.); (N.A.); (A.O.); (B.B.); (B.L.); (S.B.); (K.N.); (M.M.); (I.M.); (R.D.); (M.B.)
| |
Collapse
|
34
|
Schultheis BC, Wille C, Ross-Steinhagen NE, De Ridder D, Vancamp T, Weidle PA. Alternative Dorsal Root Ganglion Neuromodulation Electrode Implantation: A Report of 2 Cases with 3 Different Techniques. J Neurol Surg A Cent Eur Neurosurg 2021. [PMID: 34897626 DOI: 10.1055/s-0041-1739219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND STUDY AIMS The traditional percutaneous placement of dorsal root ganglion (DRG) electrodes may not be eligible for every patient. In this tertiary spine surgery and interventional pain therapy center, alternative neurostimulation implantation techniques were developed and applied where standard percutaneous approaches failed or were contraindicated. CASE PRESENTATION Three alternative implantation techniques can be used: (1) open surgical placement of DRG leads, (2) two-lead insertion via a lateral to medial transforaminal approach (level L3), and (3) percutaneous approach with two leads close to the spinal nerves L4 (peripheral nerve stimulation). RESULTS The placement of the leads occurred without complications and resulted in similar expected outcomes as with the common percutaneous technique with long-term stable pain suppression at 7 months and 1 year. CONCLUSIONS In patients in whom the DRG cannot be approached by the standard percutaneous approach, at least three alternatives may be used in experienced hands resulting in stable pain suppression of similar magnitude.
Collapse
Affiliation(s)
- Björn Carsten Schultheis
- Krankenhaus Neuwerk "Maria von den Aposteln," Muskulo-Skeletales Zentrum, interventionelle Schmerztherapie, Mönchengladbach, Germany
| | - Christian Wille
- Department of Neurosurgery, NCN Neurochirurgische Praxis Neuss, Neuss, Neuss, Germany
| | - Nikolas Eugenio Ross-Steinhagen
- Krankenhaus Neuwerk "Maria von den Aposteln," Muskulo-Skeletales Zentrum, interventionelle Schmerztherapie, Mönchengladbach, Germany
| | - Dirk De Ridder
- Department of Surgical Sciences, Section of Neurosurgery, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | | | - Patrick A Weidle
- Krankenhaus Neuwerk "Maria von den Aposteln," Muskulo-Skeletales Zentrum, interventionelle Schmerztherapie, Mönchengladbach, Germany
| |
Collapse
|
35
|
O'Connell NE, Ferraro MC, Gibson W, Rice AS, Vase L, Coyle D, Eccleston C. Implanted spinal neuromodulation interventions for chronic pain in adults. Cochrane Database Syst Rev 2021; 12:CD013756. [PMID: 34854473 PMCID: PMC8638262 DOI: 10.1002/14651858.cd013756.pub2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Implanted spinal neuromodulation (SNMD) techniques are used in the treatment of refractory chronic pain. They involve the implantation of electrodes around the spinal cord (spinal cord stimulation (SCS)) or dorsal root ganglion (dorsal root ganglion stimulation (DRGS)), and a pulse generator unit under the skin. Electrical stimulation is then used with the aim of reducing pain intensity. OBJECTIVES To evaluate the efficacy, effectiveness, adverse events, and cost-effectiveness of implanted spinal neuromodulation interventions for people with chronic pain. SEARCH METHODS We searched CENTRAL, MEDLINE Ovid, Embase Ovid, Web of Science (ISI), Health Technology Assessments, ClinicalTrials.gov and World Health Organization International Clinical Trials Registry from inception to September 2021 without language restrictions, searched the reference lists of included studies and contacted experts in the field. SELECTION CRITERIA We included randomised controlled trials (RCTs) comparing SNMD interventions with placebo (sham) stimulation, no treatment or usual care; or comparing SNMD interventions + another treatment versus that treatment alone. We included participants ≥ 18 years old with non-cancer and non-ischaemic pain of longer than three months duration. Primary outcomes were pain intensity and adverse events. Secondary outcomes were disability, analgesic medication use, health-related quality of life (HRQoL) and health economic outcomes. DATA COLLECTION AND ANALYSIS Two review authors independently screened database searches to determine inclusion, extracted data and evaluated risk of bias for prespecified results using the Risk of Bias 2.0 tool. Outcomes were evaluated at short- (≤ one month), medium- four to eight months) and long-term (≥12 months). Where possible we conducted meta-analyses. We used the GRADE system to assess the certainty of evidence. MAIN RESULTS We included 15 unique published studies that randomised 908 participants, and 20 unique ongoing studies. All studies evaluated SCS. We found no eligible published studies of DRGS and no studies comparing SCS with no treatment or usual care. We rated all results evaluated as being at high risk of bias overall. For all comparisons and outcomes where we found evidence, we graded the certainty of the evidence as low or very low, downgraded due to limitations of studies, imprecision and in some cases, inconsistency. Active stimulation versus placebo SCS versus placebo (sham) Results were only available at short-term follow-up for this comparison. Pain intensity Six studies (N = 164) demonstrated a small effect in favour of SCS at short-term follow-up (0 to 100 scale, higher scores = worse pain, mean difference (MD) -8.73, 95% confidence interval (CI) -15.67 to -1.78, very low certainty). The point estimate falls below our predetermined threshold for a clinically important effect (≥10 points). No studies reported the proportion of participants experiencing 30% or 50% pain relief for this comparison. Adverse events (AEs) The quality and inconsistency of adverse event reporting in these studies precluded formal analysis. Active stimulation + other intervention versus other intervention alone SCS + other intervention versus other intervention alone (open-label studies) Pain intensity Mean difference Three studies (N = 303) demonstrated a potentially clinically important mean difference in favour of SCS of -37.41 at short term (95% CI -46.39 to -28.42, very low certainty), and medium-term follow-up (5 studies, 635 participants, MD -31.22 95% CI -47.34 to -15.10 low-certainty), and no clear evidence for an effect of SCS at long-term follow-up (1 study, 44 participants, MD -7 (95% CI -24.76 to 10.76, very low-certainty). Proportion of participants reporting ≥50% pain relief We found an effect in favour of SCS at short-term (2 studies, N = 249, RR 15.90, 95% CI 6.70 to 37.74, I2 0% ; risk difference (RD) 0.65 (95% CI 0.57 to 0.74, very low certainty), medium term (5 studies, N = 597, RR 7.08, 95 %CI 3.40 to 14.71, I2 = 43%; RD 0.43, 95% CI 0.14 to 0.73, low-certainty evidence), and long term (1 study, N = 87, RR 15.15, 95% CI 2.11 to 108.91 ; RD 0.35, 95% CI 0.2 to 0.49, very low certainty) follow-up. Adverse events (AEs) Device related No studies specifically reported device-related adverse events at short-term follow-up. At medium-term follow-up, the incidence of lead failure/displacement (3 studies N = 330) ranged from 0.9 to 14% (RD 0.04, 95% CI -0.04 to 0.11, I2 64%, very low certainty). The incidence of infection (4 studies, N = 548) ranged from 3 to 7% (RD 0.04, 95%CI 0.01, 0.07, I2 0%, very low certainty). The incidence of reoperation/reimplantation (4 studies, N =5 48) ranged from 2% to 31% (RD 0.11, 95% CI 0.02 to 0.21, I2 86%, very low certainty). One study (N = 44) reported a 55% incidence of lead failure/displacement (RD 0.55, 95% CI 0.35, 0 to 75, very low certainty), and a 94% incidence of reoperation/reimplantation (RD 0.94, 95% CI 0.80 to 1.07, very low certainty) at five-year follow-up. No studies provided data on infection rates at long-term follow-up. We found reports of some serious adverse events as a result of the intervention. These included autonomic neuropathy, prolonged hospitalisation, prolonged monoparesis, pulmonary oedema, wound infection, device extrusion and one death resulting from subdural haematoma. Other No studies reported the incidence of other adverse events at short-term follow-up. We found no clear evidence of a difference in otherAEs at medium-term (2 studies, N = 278, RD -0.05, 95% CI -0.16 to 0.06, I2 0%) or long term (1 study, N = 100, RD -0.17, 95% CI -0.37 to 0.02) follow-up. Very limited evidence suggested that SCS increases healthcare costs. It was not clear whether SCS was cost-effective. AUTHORS' CONCLUSIONS We found very low-certainty evidence that SCS may not provide clinically important benefits on pain intensity compared to placebo stimulation. We found low- to very low-certainty evidence that SNMD interventions may provide clinically important benefits for pain intensity when added to conventional medical management or physical therapy. SCS is associated with complications including infection, electrode lead failure/migration and a need for reoperation/re-implantation. The level of certainty regarding the size of those risks is very low. SNMD may lead to serious adverse events, including death. We found no evidence to support or refute the use of DRGS for chronic pain.
Collapse
Affiliation(s)
- Neil E O'Connell
- Department of Health Sciences, Centre for Health and Wellbeing Across the Lifecourse, Brunel University London, Uxbridge, UK
| | - 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
| | - William Gibson
- School of Physiotherapy, The University of Notre Dame Australia, Fremantle, Australia
| | - Andrew Sc Rice
- Pain Research, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
| | - Lene Vase
- Department of Psychology and Behavioural Sciences, Aarhus University, Aarhus, Denmark
| | - Doug Coyle
- Epidemiology and Community Medicine, Ottawa Health Research Institute, Ottawa, Canada
- Health Economics Research Group, Institute of Environment, Health and Societies, Department of Clinical Sciences, Brunel University London, Uxbridge, UK
| | | |
Collapse
|
36
|
Chao D, Mecca CM, Yu G, Segel I, Gold MS, Hogan QH, Pan B. Dorsal root ganglion stimulation of injured sensory neurons in rats rapidly eliminates their spontaneous activity and relieves spontaneous pain. Pain 2021; 162:2917-2932. [PMID: 33990112 PMCID: PMC8486885 DOI: 10.1097/j.pain.0000000000002284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/23/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Dorsal root ganglion field stimulation (GFS) relieves evoked and spontaneous neuropathic pain by use-dependent blockade of impulse trains through the sensory neuron T-junction, which becomes complete within less than 1 minute for C-type units, also with partial blockade of Aδ units. We used this tool in the spinal nerve ligation (SNL) rat model to selectively block sensory neuron spontaneous activity (SA) of axotomized neurons at the fifth lumbar (L5) level vs blockade of units at the L4 level that remain uninjured but exposed to inflammation. In vivo dorsal root single-unit recordings after SNL showed increased SA in L5 units but not L4 units. Ganglion field stimulation blocked this SA. Ganglion field stimulation delivered at the L5 dorsal root ganglion blocked mechanical hyperalgesia behavior, mechanical allodynia, and ongoing spontaneous pain indicated by conditioned place preference, whereas GFS at L4 blocked evoked pain behavior but not spontaneous pain. In vivo single-unit recordings of spinal cord dorsal horn (DH) wide-dynamic-range neurons showed elevated SA after SNL, which was reduced by GFS at the L5 level but not by GFS at the L4 level. In addition, L5 GFS, but not L4 GFS, increased mechanical threshold of DH units during cutaneous mechanical stimulation, while L5 GFS exceeded L4 GFS in reducing evoked firing rates. Our results indicate that SA in injured neurons supports increased firing of DH wide-dynamic-range neurons, contributing to hyperalgesia, allodynia, and ongoing pain. Ganglion field stimulation analgesic effects after nerve injury are at least partly attributable to blocking propagation of this SA.
Collapse
Affiliation(s)
- Dongman Chao
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| | - Christina M. Mecca
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| | - Guoliang Yu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| | - Ian Segel
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| | - Michael S. Gold
- Department of Neurobiology, University of Pittsburgh, 3500 Terrace Street Rm E1440 BST, Pittsburgh, PA 15213
| | - Quinn H. Hogan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| | - Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226
| |
Collapse
|
37
|
Dalrymple AN, Ting JE, Bose R, Trevathan JK, Nieuwoudt S, Lempka SF, Franke M, Ludwig KA, Shoffstall AJ, Fisher LE, Weber DJ. Stimulation of the dorsal root ganglion using an Injectrode ®. J Neural Eng 2021; 18. [PMID: 34650008 DOI: 10.1088/1741-2552/ac2ffb] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/14/2021] [Indexed: 01/15/2023]
Abstract
Objective. The goal of this work was to compare afferent fiber recruitment by dorsal root ganglion (DRG) stimulation using an injectable polymer electrode (Injectrode®) and a more traditional cylindrical metal electrode.Approach. We exposed the L6 and L7 DRG in four cats via a partial laminectomy or burr hole. We stimulated the DRG using an Injectrode or a stainless steel (SS) electrode using biphasic pulses at three different pulse widths (80, 150, 300μs) and pulse amplitudes spanning the range used for clinical DRG stimulation. We recorded antidromic evoked compound action potentials (ECAPs) in the sciatic, tibial, and common peroneal nerves using nerve cuffs. We calculated the conduction velocity of the ECAPs and determined the charge-thresholds and recruitment rates for ECAPs from Aα, Aβ, and Aδfibers. We also performed electrochemical impedance spectroscopy measurements for both electrode types.Main results. The ECAP thresholds for the Injectrode did not differ from the SS electrode across all primary afferents (Aα, Aβ, Aδ) and pulse widths; charge-thresholds increased with wider pulse widths. Thresholds for generating ECAPs from Aβfibers were 100.0 ± 32.3 nC using the SS electrode, and 90.9 ± 42.9 nC using the Injectrode. The ECAP thresholds from the Injectrode were consistent over several hours of stimulation. The rate of recruitment was similar between the Injectrodes and SS electrode and decreased with wider pulse widths.Significance. The Injectrode can effectively excite primary afferents when used for DRG stimulation within the range of parameters used for clinical DRG stimulation. The Injectrode can be implanted through minimally invasive techniques while achieving similar neural activation to conventional electrodes, making it an excellent candidate for future DRG stimulation and neuroprosthetic applications.
Collapse
Affiliation(s)
- Ashley N Dalrymple
- Department of Mechanical Engineering, Carnegie Mellon University, 5000 Forbes Ave, Wean 1323, Pittsburgh, PA 15217, United States of America.,Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA, 15217, United States of America
| | - Jordyn E Ting
- Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA, 15217, United States of America.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, 15217, United States of America
| | - Rohit Bose
- Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA, 15217, United States of America.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, 15217, United States of America
| | - James K Trevathan
- Departments of Biomedical Engineering and Neurological Surgery, University of Wisconsin-Madison, Madison, WI, United States of America
| | | | - Scott F Lempka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | | | - Kip A Ludwig
- Departments of Biomedical Engineering and Neurological Surgery, University of Wisconsin-Madison, Madison, WI, United States of America.,Neuronoff Inc., Cleveland, OH, United States of America.,Wisconsin Institute for Translational Neuroengineering (WITNe), Madison, WI, United States of America
| | - Andrew J Shoffstall
- Neuronoff Inc., Cleveland, OH, United States of America.,Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States of America
| | - Lee E Fisher
- Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA, 15217, United States of America.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, 15217, United States of America.,Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Douglas J Weber
- Department of Mechanical Engineering, Carnegie Mellon University, 5000 Forbes Ave, Wean 1323, Pittsburgh, PA 15217, United States of America.,Rehab Neural Engineering Labs, University of Pittsburgh, Pittsburgh, PA, 15217, United States of America.,Center for Neural Basis of Cognition, Pittsburgh, PA, 15217, United States of America.,Neuroscience Institute, Carnegie Mellon University, 5000 Forbes Ave, Wean 1323, Pittsburgh, PA 15217, United States of America
| |
Collapse
|
38
|
Bral P, Smet I, Jerjir A, Devos M, Van Buyten JP. Dorsal root ganglion stimulation for patients with refractory pain due to anterior cutaneous nerve entrapment syndrome: A case series. Pain Pract 2021; 22:288-294. [PMID: 34672088 DOI: 10.1111/papr.13086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 08/25/2021] [Accepted: 09/25/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Anterior cutaneous nerve entrapment syndrome (ACNES) is a painful condition that can be refractory in a small percentage of patients. Abdominal pain caused by thoracic nerve entrapment in the abdominal wall characterizes ACNES. In the small number of refractory patients to all standard treatments, medication overuse and abuse are serious problems. Dorsal root ganglion (DRG) stimulation might be a good treatment to improve pain scores and to lower medication use. METHODS We describe the retrospective analysis of nine cases of patients who underwent DRG stimulation for refractory ACNES using a treatment algorithm. We focused on reported pain intensity scores, medication use, and adverse events. RESULTS All nine patients experienced a decrease in pain intensity during the trial period and received a permanent pulse generator implantation. Three months after permanent implantation, eight of nine patients showed a pain reduction of more than 50%. Medication use was substantially lower. DISCUSSION AND CONCLUSION This case series shows that DRG stimulation might be a very good treatment for refractory ACNES.
Collapse
Affiliation(s)
- Philippe Bral
- Department of Anesthesiology and Pain Medicine, UZ Brussel, Brussels, Belgium
| | - Iris Smet
- Multidisciplinary pain center, AZ Nikolaas, Sint-Niklaas, Belgium
| | - Ali Jerjir
- Multidisciplinary pain center, AZ Nikolaas, Sint-Niklaas, Belgium
| | - Marieke Devos
- Multidisciplinary pain center, AZ Nikolaas, Sint-Niklaas, Belgium
| | | |
Collapse
|
39
|
Hines K, Swaminathan V, Thalheimer S, Kogan M, Wu C, Sharan A. Single-Center Retrospective Analysis of Device-Related Complications Related to Dorsal Root Ganglion Stimulation for Pain Relief in 31 Patients. Neuromodulation 2021; 25:1040-1044. [PMID: 34309131 DOI: 10.1111/ner.13498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/06/2021] [Accepted: 06/29/2021] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Dorsal root ganglion (DRG) stimulation is a form of neuromodulation used to treat neuropathic pain due to a myriad of etiologies. Though this relatively new therapy has been shown to be quite effective, complications associated with the implantation of this therapy have not been well documented. OBJECTIVES The primary objective of this study was to describe the device-related complications associated with DRG stimulator implantations. MATERIALS AND METHODS This was a single-center retrospective analysis of 31 patients who underwent full implantation of neuromodulation hardware marketed for DRG stimulation. The predefined endpoints included device-related complications associated with DRG implantations, such as hardware failure, explantation procedures, and revision surgery. Additional endpoints included percentage of patients receiving therapy and pain as measured using the visual analog scale (VAS) pain scale at initial, six-month, and 12-month follow-up after hardware implantation. RESULTS Thirty-one patients were included out of 42 patients trialed. Baseline VAS in patients was 7.7 (31 patients). At initial follow-up, six-month follow-up, and one-year follow-up, VAS scores were 4.7 (31 patients), 5.3 (20 patients), and 5.5 (13 patients), respectively. Paired t-test between preoperative VAS (mean 7.3) and one-year follow-up VAS (5.5) demonstrated statistical significance (p = 0.027). At initial, six-month, and one-year follow-up, 30/31 (97%), 19/24 (79%), and 18/23 (78%) patients were confirmed to be receiving DRG stimulation therapy after permanent implant. Of the 31 patients who were implanted with a permanent system, 8 (26%) were explanted and an additional 10 (29%) required revision surgery. CONCLUSION In this study, we examine the various device-related complications associated with DRG stimulation requiring repeat surgery. High rates of hardware failure, revision surgery, and explantation of stimulators illustrate the need for hardware optimization to improve patient outcomes.
Collapse
Affiliation(s)
- Kevin Hines
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Vishal Swaminathan
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sara Thalheimer
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael Kogan
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Chengyuan Wu
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ashwini Sharan
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
40
|
Yu G, Segel I, Tran H, Park HJ, Ross E, Hogan QH, Pan B. Analgesic Effects of Tonic and Burst Dorsal Root Ganglion Stimulation in Rats With Painful Tibial Nerve Injury. Neuromodulation 2021; 25:970-979. [PMID: 34096146 DOI: 10.1111/ner.13472] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/06/2021] [Accepted: 05/17/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Dorsal root ganglion (DRG) stimulation is effective in treating chronic pain. While burst stimulation has been proven to enhance the therapeutic efficacy in spinal cord stimulation, currently only a tonic stimulation waveform is clinically used in DRG stimulation. We hypothesized that burst DRG stimulation might also produce analgesic effect in a preclinical neuropathic pain model. We evaluated both the therapeutic effects of burst DRG stimulation and the possible effects of DRG stimulation upon inflammation within the DRG in a preclinical neuropathic pain model. MATERIALS AND METHODS Rats received either a painful tibial nerve injury or sham surgery. Analgesic effects of DRG stimulation were evaluated by testing a battery of evoked pain-related behaviors as well as measuring the positive affective state associated with relief of spontaneous pain using conditioned place preference. Histological evidence for neuronal trauma or neuroinflammation was evaluated. RESULTS All of the waveforms tested (20 Hz-tonic, 20 Hz-burst, and 40 Hz-burst) have similar analgesic effects in sensory tests and conditioned place preference. Long-term DRG stimulation for two weeks does not change DRG expression of markers for nerve injury and neuroinflammation. CONCLUSIONS DRG stimulation using burst waveform might be also suitable for treating neuropathic pain.
Collapse
Affiliation(s)
- Guoliang Yu
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Ian Segel
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hai Tran
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | | | - Erika Ross
- Abbott Neuromodulation, Plano, TX, 75024, USA
| | - Quinn H Hogan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Bin Pan
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| |
Collapse
|
41
|
Soloukey S, de Rooij JD, Osterthun R, Drenthen J, De Zeeuw CI, Huygen FJPM, Harhangi BS. The Dorsal Root Ganglion as a Novel Neuromodulatory Target to Evoke Strong and Reproducible Motor Responses in Chronic Motor Complete Spinal Cord Injury: A Case Series of Five Patients. Neuromodulation 2021; 24:779-793. [PMID: 32706445 PMCID: PMC8359424 DOI: 10.1111/ner.13235] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Current strategies for motor recovery after spinal cord injury (SCI) aim to facilitate motor performance through modulation of afferent input to the spinal cord using epidural electrical stimulation (EES). The dorsal root ganglion (DRG) itself, the first relay station of these afferent inputs, has not yet been targeted for this purpose. The current study aimed to determine whether DRG stimulation can facilitate clinically relevant motor response in motor complete SCI. MATERIALS AND METHODS Five patients with chronic motor complete SCI were implanted with DRG leads placed bilaterally on level L4 during five days. Based on personalized stimulation protocols, we aimed to evoke dynamic (phase 1) and isotonic (phase 2) motor responses in the bilateral quadriceps muscles. On days 1 and 5, EMG-measurements (root mean square [RMS] values) and clinical muscle force measurements (MRC scoring) were used to measure motor responses and their reproducibility. RESULTS In all patients, DRG-stimulation evoked significant phase 1 and phase 2 motor responses with an MRC ≥4 for all upper leg muscles (rectus femoris, vastus lateralis, vastus medialis, and biceps femoris) (p < 0.05 and p < 0.01, respectively), leading to a knee extension movement strong enough to facilitate assisted weight bearing. No significant differences in RMS values were observed between days 1 and 5 of the study, indicating that motor responses were reproducible. CONCLUSION The current paper provides first evidence that bilateral L4 DRG stimulation can evoke reproducible motor responses in the upper leg, sufficient for assisted weight bearing in patients with chronic motor complete SCI. As such, a new target for SCI treatment has surfaced, using existing stimulation devices, making the technique directly clinically accessible.
Collapse
Affiliation(s)
- Sadaf Soloukey
- Department of NeurosurgeryErasmus MC RotterdamRotterdamThe Netherlands
- Department of NeuroscienceErasmus MC RotterdamRotterdamThe Netherlands
| | - Judith D. de Rooij
- Center for Pain Medicine, Department of AnesthesiologyErasmus MC RotterdamRotterdamThe Netherlands
- Unit of Physiotherapy, Department of OrthopedicsErasmus MC RotterdamRotterdamThe Netherlands
| | - Rutger Osterthun
- Department of Rehabilitation MedicineErasmus MC RotterdamRotterdamThe Netherlands
- Spinal Cord Injury DepartmentRijndam Rehabilitation CenterRotterdamThe Netherlands
| | - Judith Drenthen
- Department of Clinical NeurophysiologyErasmus MC RotterdamRotterdamThe Netherlands
| | - Chris I. De Zeeuw
- Department of NeuroscienceErasmus MC RotterdamRotterdamThe Netherlands
- Netherlands Institute for NeuroscienceRoyal Dutch Academy for Arts and SciencesAmsterdamThe Netherlands
| | - Frank J. P. M. Huygen
- Center for Pain Medicine, Department of AnesthesiologyErasmus MC RotterdamRotterdamThe Netherlands
| | | |
Collapse
|
42
|
Analgesic dorsal root ganglionic field stimulation blocks conduction of afferent impulse trains selectively in nociceptive sensory afferents. Pain 2021; 161:2872-2886. [PMID: 32658148 DOI: 10.1097/j.pain.0000000000001982] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Increased excitability of primary sensory neurons after peripheral nerve injury may cause hyperalgesia and allodynia. Dorsal root ganglion field stimulation (GFS) is effective in relieving clinical pain associated with nerve injury and neuropathic pain in animal models. However, its mechanism has not been determined. We examined effects of GFS on transmission of action potentials (APs) from the peripheral to central processes by in vivo single-unit recording from lumbar dorsal roots in sham injured rats and rats with tibial nerve injury (TNI) in fiber types defined by conduction velocity. Transmission of APs directly generated by GFS (20 Hz) in C-type units progressively abated over 20 seconds, whereas GFS-induced Aβ activity persisted unabated, while Aδ showed an intermediate pattern. Activity generated peripherally by electrical stimulation of the sciatic nerve and punctate mechanical stimulation of the receptive field (glabrous skin) was likewise fully blocked by GFS within 20 seconds in C-type units, whereas Aβ units were minimally affected and a subpopulation of Aδ units was blocked. After TNI, the threshold to induce AP firing by punctate mechanical stimulation (von Frey) was reduced, which was reversed to normal during GFS. These results also suggest that C-type fibers, not Aβ, mainly contribute to mechanical and thermal hypersensitivity (von Frey, brush, acetone) after injury. Ganglion field stimulation produces use-dependent blocking of afferent AP trains, consistent with enhanced filtering of APs at the sensory neuron T-junction, particularly in nociceptive units.
Collapse
|
43
|
Nagpal A, Clements N, Duszynski B, Boies B. The Effectiveness of Dorsal Root Ganglion Neurostimulation for the Treatment of Chronic Pelvic Pain and Chronic Neuropathic Pain of the Lower Extremity: A Comprehensive Review of the Published Data. PAIN MEDICINE 2021; 22:49-59. [PMID: 33260203 DOI: 10.1093/pm/pnaa369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To evaluate the effectiveness of dorsal root ganglion neurostimulation for the treatment of refractory, focal pain in the pelvis and lower extremities. DESIGN Systematic review. OUTCOME MEASURES The primary outcome was ≥50% pain relief. Secondary outcomes were physical function, mood, quality of life, opioid usage, and complications. RESULTS One pragmatic randomized controlled trial, four prospective cohort studies, and eight case series met the inclusion criteria. A worst-case scenario analysis from the randomized controlled trial reported ≥50% pain relief in 74% of patients with dorsal root ganglion neurostimulation vs. 51% of patients who experienced at least 50% relief with spinal cord stimulation at 3 months. Cohort data success rates ranged from 43% to 83% at ≤6 months and 27% to 100% at >6 months. Significant improvements were also reported in the secondary outcomes assessed, including mood, quality of life, opioid usage, and health care utilization, though a lack of available quantitative data limits further statistical analysis. Complication rates vary, though the only randomized controlled trial reported a higher rate of adverse events than that seen with traditional neurostimulation. CONCLUSIONS In accordance with the Grades of Recommendation, Assessment, Development, and Evaluation system, low-quality evidence supports dorsal root ganglion neurostimulation as a more effective treatment than traditional neurostimulation for pain and dysfunction associated with complex regional pain syndrome or causalgia. Very low-quality evidence supports dorsal root ganglion neurostimulation for the treatment of chronic pelvic pain, chronic neuropathic groin pain, phantom limb pain, chronic neuropathic pain of the trunk and/or limbs, and diabetic neuropathy.
Collapse
Affiliation(s)
- Ameet Nagpal
- Department of Anesthesiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Nathan Clements
- Department of Rehabilitation Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | | | - Brian Boies
- Department of Anesthesiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
44
|
Interventional Therapies for Pain in Cancer Patients: a Narrative Review. Curr Pain Headache Rep 2021; 25:44. [PMID: 33961156 DOI: 10.1007/s11916-021-00963-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE OF REVIEW Pain is a prevalent symptom in the lives of patients with cancer. In light of the ongoing opioid epidemic and increasing awareness of the potential for opioid abuse and addiction, clinicians are progressively turning to interventional therapies. This article reviews the interventional techniques available to mitigate the debilitating effects that untreated or poorly treated pain have in this population. RECENT FINDINGS A range of interventional therapies and technical approaches are available for the treatment of cancer-related pain. Many of the techniques described may offer effective analgesia with less systemic toxicity and dependency than first- and second-line oral and parenteral agents. Neuromodulatory techniques including dorsal root ganglion stimulation and peripheral nerve stimulation are increasingly finding roles in the management of oncologic pain. The goal of this pragmatic narrative review is to discuss interventional approaches to cancer-related pain and the potential of such therapies to improve the quality of life of cancer patients.
Collapse
|
45
|
Spinal cord stimulation in chronic neuropathic pain: mechanisms of action, new locations, new paradigms. Pain 2021; 161 Suppl 1:S104-S113. [PMID: 33090743 PMCID: PMC7434213 DOI: 10.1097/j.pain.0000000000001854] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
46
|
Berger AA, Liu Y, Possoit H, Rogers AC, Moore W, Gress K, Cornett EM, Kaye AD, Imani F, Sadegi K, Varrassi G, Viswanath O, Urits I. Dorsal Root Ganglion (DRG) and Chronic Pain. Anesth Pain Med 2021; 11:e113020. [PMID: 34336621 PMCID: PMC8314073 DOI: 10.5812/aapm.113020] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
CONTEXT Chronic neuropathic pain is a common condition, and up to 11.9% of the population have been reported to suffer from uncontrolled neuropathic pain. Chronic pain leads to significant morbidity, lowered quality of life, and loss of workdays, and thus carries a significant price tag in healthcare costs and lost productivity. dorsal root ganglia (DRG) stimulation has been recently increasingly reported and shows promising results in the alleviation of chronic pain. This paper reviews the background of DRG stimulation, anatomical, and clinical consideration and reviews the clinical evidence to support its use. EVIDENCE ACQUISITION The DRG span the length of the spinal cord and house the neurons responsible for sensation from the periphery. They may become irritated by direct compression or local inflammation. Glial cells in the DRG respond to nerve injury, producing inflammatory markers and contribute to the development of chronic pain, even after the resolution of the original insult. While the underlying mechanism is still being explored, recent studies explored the efficacy of DRG stimulation and neuromodulation for chronic pain treatment. RESULTS Several reported cases and a small number of randomized trials were published in recent years, describing different methods of DRG stimulation and neuromodulation with promising results. Though evidence quality is mostly low, these results provide evidence to support the utilization of this technique. CONCLUSIONS Chronic neuropathic pain is a common condition and carries significant morbidity and impact on the quality of life. Recent evidence supports the use of DRG neuromodulation as an effective technique to control chronic pain. Though studies are still emerging, the evidence appears to support this technique. Further studies, including large randomized trials evaluating DRG modulation versus other interventional and non-interventional techniques, are needed to further elucidate the efficacy of this method. These studies are also likely to inform the patient selection and the course of treatment.
Collapse
Affiliation(s)
- Amnon A. Berger
- Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA, USA
| | - Yao Liu
- Beth Israel Deaconess Medical Center, Department of Anesthesiology, Critical Care, and Pain Medicine, Harvard Medical School, Boston, MA, USA
| | - HarLee Possoit
- LSU Health Shreveport, School of Medicine, Shreveport, LA, USA
| | - Anna C. Rogers
- LSU Health Shreveport, School of Medicine, Shreveport, LA, USA
| | - Warner Moore
- LSU Health Shreveport, School of Medicine, Shreveport, LA, USA
| | - Kyle Gress
- Georgetown University School of Medicine, Washington, DC, USA
| | - Elyse M. Cornett
- LSU Health Shreveport, Department of Anesthesiology, Shreveport, LA, USA
| | - Alan David Kaye
- LSU Health Shreveport, Department of Anesthesiology, Shreveport, LA, USA
| | - Farnad Imani
- Pain Research Center, Department of Anesthesiology and Pain Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kambiz Sadegi
- Department of Anesthesiology, Zabol University of Medical Sciences, Zabol, Iran
| | | | - Omar Viswanath
- Georgetown University School of Medicine, Washington, DC, USA
- University of Arizona College of Medicine - Phoenix, Department of Anesthesiology, Phoenix, AZ, USA
- Creighton University School of Medicine, Department of Anesthesiology, Omaha, NE, USA
- Valley Anesthesiology and Pain Consultants – Envision Physician Services, Phoenix, AZ, USA
| | - Ivan Urits
- Georgetown University School of Medicine, Washington, DC, USA
- Southcoast Health, Southcoast Health Physicians Group Pain Medicine, Wareham, MA, USA
| |
Collapse
|
47
|
Grabnar M, Kim C. Dorsal Root Ganglion Stimulation for Treatment of Chemotherapy-Induced Neuropathy. Am J Phys Med Rehabil 2021; 100:e52-e54. [PMID: 32701635 DOI: 10.1097/phm.0000000000001542] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Chemotherapy-induced neuropathy has limited treatment options, and conventional medications used to treat neuropathic pain often do not provide adequate pain relief for patients with a history of cancer. Neuromodulation such as dorsal root ganglion stimulation remains a treatment that has been studied for chronic painful conditions such as low back pain, pelvic pain, complex regional pain syndrome, and phantom limb pain. Dorsal root ganglion stimulation has been presented for treatment of chemotherapy-induced neuropathy, but with limited duration of follow-up. We present a case of pain resolution after placement of a dorsal root ganglion stimulation for persistent chemotherapy-induced neuropathy. Our patient developed burning pain and allodynia in both feet 3 mos into her chemotherapy regimen, with worsened symptoms after cessation of chemotherapy. After failure of conservative pharmacotherapies, a 7-day dorsal root ganglion stimulation trial was implanted, resulting in 100% pain relief. A dorsal root ganglion stimulation was then implanted permanently, and our patient reported continued resolution of symptoms at evaluation 3 yrs after placement. To the author's knowledge, this is the first case of sustained relief with dorsal root ganglion stimulation placement for chemotherapy-induced neuropathy and presents a treatment option that warrants further investigation.
Collapse
Affiliation(s)
- Maria Grabnar
- From the Case Western Reserve University/MetroHealth, Cleveland, Ohio
| | | |
Collapse
|
48
|
Prasad Md A, Chakravarthy Md K. Review of complex regional pain syndrome and the role of the neuroimmune axis. Mol Pain 2021; 17:17448069211006617. [PMID: 33788654 PMCID: PMC8020088 DOI: 10.1177/17448069211006617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background Complex regional pain syndrome (CRPS) is a progressive and painful disease of
the extremities that is characterized by continuous pain inconsistent with
the initial trauma. CRPS is caused by a multi-mechanism process that
involves both the peripheral and central nervous system, with a prominent
role of inflammation in CRPS pathophysiology. This review examines what is
currently known about the CRPS inflammatory and pain mechanisms, as well as
the possible impact of neurostimulation therapies on the neuroimmune axis of
CRPS. Study design A narrative review of preclinical and clinical studies provided an overview
of the pain and inflammatory mechanisms in CRPS and addressed the effect of
neurostimulation on immunomodulation. Methods A systematic literature search was conducted based on the PRISMA guidelines
between September 2015 to September 2020. Data sources included relevant
literature identified through searches of PubMed, Embase and the Cochrane
Database of Systematic Reviews. Results Sixteen preclinical and eight clinical studies were reviewed. Preclinical
studies identified different mechanisms of pain development in the acute and
chronic CRPS phases. Several preclinical and clinical studies investigating
inflammatory mechanisms, autoimmunity, and genetic profiles in CRPS,
supported a role of neuroinflammation in the pathophysiology of CRPS. The
immunomodulatory effects of neurostimulation therapy is still unclear,
despite clinical improvement in the CRPS patients. Conclusions Increasing evidence supports a role for inflammation and neuroinflammation in
CRPS pathophysiology. Preliminary neurostimulation findings, together with
the role of (neuro)inflammation in CRPS, seems to provide a compelling
rationale for its use in CRPS pain treatment. The possible immunomodulatory
effects of neurostimulation opens new therapeutic possibilities, however
further research is needed to gain a better understanding of the working
mechanisms.
Collapse
Affiliation(s)
- Amrita Prasad Md
- Axxon Pain, Brisbane Private Hospital, 259 Wickham Terrace, Brisbane, Queensland 4000, Australia
| | - Krishnan Chakravarthy Md
- Division of Pain Medicine, Department of Anesthesiology, University of California San Diego, La Jolla, CA, USA.,Department of Anesthesiology and Pain Medicine, VA San Diego Health Care, San Diego, CA, USA
| |
Collapse
|
49
|
Stelter B, Karri J, Marathe A, Abd-Elsayed A. Dorsal Root Ganglion Stimulation for the Treatment of Non-Complex Regional Pain Syndrome Related Chronic Pain Syndromes: A Systematic Review. Neuromodulation 2021; 24:622-633. [PMID: 33501749 DOI: 10.1111/ner.13361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/08/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND While the majority of indications and approvals for dorsal root ganglion stimulation (DRGS) are for the refractory management of complex regional pain syndrome (CRPS), emerging evidence has suggested that DRGS may be favorably used for a plethora of other chronic pain phenomena. Consequently, we aimed to characterize the use and efficacy of DRGS for these non-CRPS-related chronic pain syndromes. MATERIALS AND METHODS A systematic review of clinical studies demonstrating the use of DRGS for non-CRPS-related chronic pain syndromes. The literature search was performed using PubMed, Cochrane Library, and CINAHL plus across August and September 2020. RESULTS A total of 28 reports comprising 354 total patients were included in the analysis. Of the chronic pain syndromes presented, axial low back pain, chronic pelvic and groin pain, other peripheral neuropathies, and studies with multiple concomitant pain syndromes, a majority demonstrated >50% mean pain reduction at the time of last follow-up following DRGS. Physical function, quality of life (QOL), and lesser pain medication usage also were repeatedly reported to be significantly improved. CONCLUSIONS DRGS continues to lack supportive evidence from well designed, high level studies and recommendations from consensus committee experts. However, we present repeated and consistent evidence from lower level studies showing success with the use of DRGS for various non-CRPS chronic pain syndromes in reducing pain along with increasing function and QOL from one week to three years. Due to such low-level, high bias evidence, we strongly encourage the continuation of high-level studies in order to provide a stronger foundation for the use of DRGS in non-CRPS chronic pain patients. However, it may be reasonable and appropriate to evaluate patients for DRGS candidacy on a case-by-case basis particularly if they manifest focal pain syndromes refractory to noninterventional measures and may not be ideal candidates for other forms of neuromodulation.
Collapse
Affiliation(s)
- Bradly Stelter
- Department of Anesthesia, Division of Pain Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jay Karri
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Anuj Marathe
- Department of Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Alaa Abd-Elsayed
- Department of Anesthesia, Division of Pain Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
50
|
Sankarasubramanian V, Chiravuri S, Mirzakhalili E, Anaya CJ, Scott JR, Brummett CM, Clauw DJ, Patil PG, Harte SE, Lempka SF. Quantitative Sensory Testing of Spinal Cord and Dorsal Root Ganglion Stimulation in Chronic Pain Patients. Neuromodulation 2021; 24:672-684. [PMID: 33471409 DOI: 10.1111/ner.13329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/17/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES The physiological mechanisms underlying the pain-modulatory effects of clinical neurostimulation therapies, such as spinal cord stimulation (SCS) and dorsal root ganglion stimulation (DRGS), are only partially understood. In this pilot prospective study, we used patient-reported outcomes (PROs) and quantitative sensory testing (QST) to investigate the physiological effects and possible mechanisms of action of SCS and DRGS therapies. MATERIALS AND METHODS We tested 16 chronic pain patients selected for SCS and DRGS therapy, before and after treatment. PROs included pain intensity, pain-related symptoms (e.g., pain interference, pain coping, sleep interference) and disability, and general health status. QST included assessments of vibration detection theshold (VDT), pressure pain threshold (PPT) and tolerance (PPToL), temporal summation (TS), and conditioned pain modulation (CPM), at the most painful site. RESULTS Following treatment, all participants reported significant improvements in PROs (e.g., reduced pain intensity [p < 0.001], pain-related functional impairment [or pain interference] and disability [p = 0.001 for both]; better pain coping [p = 0.03], sleep [p = 0.002]), and overall health [p = 0.005]). QST showed a significant treatment-induced increase in PPT (p = 0.002) and PPToL (p = 0.011), and a significant reduction in TS (p = 0.033) at the most painful site, but showed no effects on VDT and CPM. We detected possible associations between a few QST measures and a few PROs. Notably, higher TS was associated with increased pain interference scores at pre-treatment (r = 0.772, p = 0.009), and a reduction in TS was associated with the reduction in pain interference (r = 0.669, p = 0.034) and pain disability (r = 0.690, p = 0.027) scores with treatment. CONCLUSIONS Our preliminary findings suggest significant clinical and therapeutic benefits associated with SCS and DRGS therapies, and the possible ability of these therapies to modulate pain processing within the central nervous system. Replication of our pilot findings in future, larger studies is necessary to characterize the physiological mechanisms of SCS and DRGS therapies.
Collapse
Affiliation(s)
- Vishwanath Sankarasubramanian
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Srinivas Chiravuri
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Ehsan Mirzakhalili
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Carlos J Anaya
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - John Ryan Scott
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Chad M Brummett
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel J Clauw
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA.,Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Parag G Patil
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA.,Department of Neurological Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Steven E Harte
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA.,Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Scott F Lempka
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.,Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.,Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|