Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 225
Filter
1.
Curr Pain Headache Rep ; 28(9): 941-947, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38850491

ABSTRACT

PURPOSE OF REVIEW: Spinal cord stimulation (SCS) is an increasingly utilized therapy for the treatment of neuropathic pain conditions. Though minimally invasive and reversable, there are several important device-related complications that physicians should be aware of before offering this therapy to patients. The aim of this review is to synthesize recent studies in device-related SCS complications pertaining to cylindrical lead implantation and to discuss etiologies, symptoms and presentations, diagnostic evaluation, clinical implications, and treatment options. RECENT FINDINGS: Device-related complications are more common than biologic complications. Device-related complications covered in this review include lead migration, lead fracture, lead disconnection, generator failure, loss of charge, generator flipping, hardware related pain, and paresthesia intolerance. The use of SCS continues to be an effective option for neuropathic pain conditions. Consideration of complications prior to moving forward with SCS trials and implantation is a vital part of patient management and device selection. Knowledge of these complications can provide physicians and other healthcare professionals the ability to maximize patient outcomes.


Subject(s)
Neuralgia , Spinal Cord Stimulation , Humans , Spinal Cord Stimulation/adverse effects , Spinal Cord Stimulation/instrumentation , Spinal Cord Stimulation/methods , Neuralgia/therapy , Neuralgia/etiology , Equipment Failure , Electrodes, Implanted/adverse effects
2.
Neuron ; 112(3): 331-333, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38330897

ABSTRACT

Neurostimulation produces unnatural cutaneous sensations with potent analgesic effects in pain syndromes. In this issue of Neuron, Sagalajev et al.1 demonstrate that these sensations are an epiphenomenon and explain how high-frequency stimulation can provide analgesia without these unnecessary sensations.


Subject(s)
Paresthesia , Spinal Cord Stimulation , Humans , Paresthesia/therapy , Paresthesia/etiology , Pain Measurement , Pain/complications , Pain Management , Axons/physiology , Spinal Cord Stimulation/adverse effects
3.
Pain Pract ; 24(1): 91-100, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37626446

ABSTRACT

BACKGROUND: Neurologic deficit is known as a rare complication of thoracic spinal cord stimulator (SCS) paddle lead implantation, but many believe its incidence after SCS paddle lead placement is under-reported. It is possible that imaging characteristics may be used to help predict safe paddle lead placement. OBJECTIVE: This imaging study was undertaken to determine the minimum canal diameter required for safe paddle lead placement. METHODS: Patients who underwent thoracic laminotomy for new SCS paddle lead placement from January 2018 to March 2023 were identified retrospectively. Preoperative thoracic canal diameter was measured in the sagittal plane perpendicular to the disc space from T5/6 to T11/12. These thoracic levels were chosen because they span the most common levels targeted for SCS placement. Patients with and without new neurologic deficits were compared using a Mann-Whitney U-test. RESULTS: Of 185 patients initially identified, 180 had thoracic imaging available for review. One (0.5%) and 2 (1.1%) of 185 patients complained of permanent and transient neurologic deficit after thoracic SCS placement, respectively. Patients with neurologic deficits had average canal diameters of <11 mm. The average canal diameter of patients with and without neurologic deficits was 10.2 mm (range 6.1-12.9 mm) and 13.0 mm (range 5.9-20.2), respectively (p < 0.0001). CONCLUSION: Postoperative neurologic deficit is an uncommon complication after thoracic laminotomy for SCS paddle lead placement. The authors recommend ensuring a starting thoracic canal diameter of at least 12 mm to accommodate a SCS paddle lead measuring 2 mm thick to ensure a final diameter of >10 mm. If canal diameter is <12 mm, aggressive undercutting of the lamina, a second laminotomy, or placement of smaller SCS wire leads should be considered.


Subject(s)
Electric Stimulation Therapy , Spinal Cord Stimulation , Humans , Retrospective Studies , Electrodes, Implanted/adverse effects , Spinal Cord , Electric Stimulation Therapy/methods , Spinal Canal/diagnostic imaging , Spinal Canal/surgery , Spinal Cord Stimulation/adverse effects , Spinal Cord Stimulation/methods
4.
Pain Pract ; 24(2): 308-320, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37859565

ABSTRACT

INTRODUCTION: Pain as a symptom of diabetic polyneuropathy (DPN) significantly lowers quality of life, increases mortality and is the main reason for patients with diabetes to seek medical attention. The number of people suffering from painful diabetic polyneuropathy (PDPN) has increased significantly over the past decades. METHODS: The literature on the diagnosis and treatment of diabetic polyneuropathy was retrieved and summarized. RESULTS: The etiology of PDPN is complex, with primary damage to peripheral nociceptors and altered spinal and supra-spinal modulation. To achieve better patient outcomes, the mode of diagnosis and treatment of PDPN evolves toward more precise pain-phenotyping and genotyping based on patient-specific characteristics, new diagnostic tools, and prior response to pharmacological treatments. According to the Toronto Diabetic Neuropathy Expert Group, a presumptive diagnosis of "probable PDPN" is sufficient to initiate treatment. Proper control of plasma glucose levels, and prevention of risk factors are essential in the treatment of PDPN. Mechanism-based pharmacological treatment should be initiated as early as possible. If symptomatic pharmacologic treatment fails, spinal cord stimulation (SCS) should be considered. In isolated cases, where symptomatic pharmacologic treatment and SCS are unsuccessful or cannot be used, sympathetic lumbar chain neurolysis and/or radiofrequency ablation (SLCN/SLCRF), dorsal root ganglion stimulation (DRGs) or posterior tibial nerve stimulation (PTNS) may be considered. However, it is recommended that these treatments be applied only in a study setting in a center of expertise. CONCLUSIONS: The diagnosis of PDPN evolves toward pheno-and genotyping and treatment should be mechanism-based.


Subject(s)
Diabetes Mellitus , Diabetic Neuropathies , Spinal Cord Stimulation , Humans , Diabetic Neuropathies/diagnosis , Diabetic Neuropathies/therapy , Diabetic Neuropathies/complications , Pain Management/adverse effects , Quality of Life , Pain Measurement/adverse effects , Pain/etiology , Spinal Cord Stimulation/adverse effects
5.
Pain Pract ; 24(4): 584-599, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38078593

ABSTRACT

INTRODUCTION: The use of spinal cord stimulation (SCS) therapy to treat chronic pain continues to rise. Optimal patient selection remains one of the most important factors for SCS success. However, despite increased utilization and the existence of general indications, predicting which patients will benefit from neuromodulation remains one of the main challenges for this therapy. Therefore, this study aims to identify the variables that may correlate with nonresponders to high-frequency (10 kHz) SCS to distinguish the subset of patients less likely to benefit from this intervention. MATERIALS AND METHODS: This was a retrospective single-center observational study of patients who underwent 10 kHz SCS implant. Patients were divided into nonresponders and responders groups. Demographic data and clinical outcomes were collected at baseline and statistical analysis was performed for all continuous and categorical variables between the two groups to calculate statistically significant differences. RESULTS: The study population comprised of 237 patients, of which 67.51% were responders and 32.49% were nonresponders. There was a statistically significant difference of high levels of kinesiophobia, high self-perceived disability, greater pain intensity, and clinically relevant pain catastrophizing at baseline in the nonresponders compared to the responders. A few variables deemed potentially relevant, such as age, gender, history of spinal surgery, diabetes, alcohol use, tobacco use, psychiatric illness, and opioid utilization at baseline were not statistically significant. CONCLUSION: Our study is the first in the neuromodulation literature to raise awareness to the association of high levels of kinesiophobia preoperatively in nonresponders to 10 kHz SCS therapy. We also found statistically significant differences with greater pain intensity, higher self-perceived disability, and clinically relevant pain catastrophizing at baseline in the nonresponders relative to responders. It may be appropriate to screen for these factors preoperatively to identify patients who are less likely to respond to SCS. If these modifiable risk factors are present, it might be prudent to consider a pre-rehabilitation program with pain neuroscience education to address these factors prior to SCS therapy, to enhance successful outcomes in neuromodulation.


Subject(s)
Chronic Pain , Spinal Cord Stimulation , Humans , Spinal Cord Stimulation/adverse effects , Retrospective Studies , Chronic Pain/therapy , Chronic Pain/etiology , Pain Management , Catastrophization , Treatment Outcome , Spinal Cord
6.
Stereotact Funct Neurosurg ; 102(1): 13-23, 2024.
Article in English | MEDLINE | ID: mdl-38052193

ABSTRACT

INTRODUCTION: Neuromodulation using deep brain stimulation (DBS), spinal cord stimulation (SCS), and peripheral nerve field stimulation (PNFS) to treat neurological, psychiatric, and pain disorders is a rapidly growing field. Infections related to the implanted hardware are among the most common complications and result in health-related and economic burden. Unfortunately, conservative medical therapy is less likely to be successful. In this retrospective study, we aimed to identify characteristics of the infections and investigated surgical and antimicrobial treatments. METHODS: A retrospective analysis was performed of patients with an infection related to DBS, SCS, and/or PNFS hardware over an 8-year period at our institution. Data were analyzed for type of neurostimulator, time of onset of infection following the neurosurgical procedure, location, and surgical treatment strategy. Surgical treatment of infections consisted of either a surgical wound revision without hardware removal or a surgical wound revision with partial or complete hardware removal. Data were further analyzed for the microorganisms involved, antimicrobial treatment and its duration, and clinical outcome. RESULTS: Over an 8-year period, a total of 1,250 DBS, 1,835 SCS, and 731 PNFS surgeries were performed including de novo system implantations, implanted pulse generator (IPG) replacements, and revisions. We identified 82 patients with infections related to the neurostimulator hardware, representing an incidence of 3.09% of the procedures. Seventy-one percent of the patients had undergone multiple surgeries related to the neurostimulator prior to the infection. The infections occurred after a mean of 12.2 months after the initial surgery. The site of infection was most commonly around the IPG, especially in DBS and SCS. The majority (62.2%) was treated by surgical wound revision with simultaneous partial or complete removal of hardware. Microbiological specimens predominantly yielded Staphylococcus epidermidis (39.0%) and Staphylococcus aureus (35.4%). After surgery, antimicrobials were given for a mean of 3.4 weeks. The antimicrobial regime was significantly shorter in patients with hardware removal in comparison to those who only had undergone surgical wound revision. One intracranial abscess occurred. No cases of infection-related death, sepsis, bacteremia, or intraspinal abscesses were found. CONCLUSION: Our data did show the predominance of S. epidermidis and S. aureus as etiologic organisms in hardware-related infections. Infections associated with S. aureus most likely required (partial) hardware removal. Aggressive surgical treatment including hardware removal shortens the duration of antimicrobial treatment. Clear strategies should be developed to treat hardware-related infections to optimize patient management and reduce health- and economic-related burden.


Subject(s)
Deep Brain Stimulation , Spinal Cord Stimulation , Surgical Wound , Humans , Incidence , Retrospective Studies , Staphylococcus aureus , Deep Brain Stimulation/adverse effects , Deep Brain Stimulation/methods , Surgical Wound/drug therapy , Surgical Wound Infection/epidemiology , Surgical Wound Infection/etiology , Surgical Wound Infection/surgery , Anti-Bacterial Agents , Spinal Cord , Spinal Cord Stimulation/adverse effects , Electrodes, Implanted/adverse effects
7.
Curr Pain Headache Rep ; 28(1): 1-9, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37855944

ABSTRACT

PURPOSE OF REVIEW: Spinal cord stimulation has been increasing in influence as an option to regulate pain, especially in the chronic pain patient population. However, even with the numerous changes made to this technology since its inception, it is still prone to various complications such as hardware issues, neurological injury/epidural hematoma, infections, and other biological concerns. The purpose of this article is to thoroughly review and evaluate literature pertaining to the complications associated with percutaneous spinal cord stimulation. RECENT FINDINGS: Lead migration is generally the most common complication of percutaneous spinal cord stimulation; however, recent utilization of various anchoring techniques has been discussed and experienced clinical success in decreasing the prevalence of lead migration and lead fractures. With newer high-frequency systems gaining traction to improve pain management and decrease complications as compared to traditional systems, rechargeable implantable pulse generators have been the preferred power source. However, recent findings may suggest that these rechargeable implantable pulse generators do not significantly increase battery life as much as was proposed. Intraoperative neuromonitoring has seen success in mitigating neurological injury postoperatively and may see more usage in the future through more testing. Though the occurrence of infection and biological complications, including dural puncture and skin erosion, has been less frequent over time, they should still be treated in accordance with established protocols. While many complications can arise following percutaneous spinal cord stimulator implantation, the procedure is less invasive than open implantation and has seen largely positive patient feedback. Hardware complications, the more common issues that can occur, rarely indicate a serious risk and can generally be remedied through reoperation. However, less common cases such as neurological injury, infections, and biological complications require prompt diagnosis to improve the condition of the patient and prevent significant damage.


Subject(s)
Spinal Cord Stimulation , Humans , Electrodes, Implanted/adverse effects , Spinal Cord Stimulation/adverse effects , Spinal Cord Stimulation/methods , Pain Management/methods , Neurosurgical Procedures/methods , Spinal Cord
8.
Reg Anesth Pain Med ; 49(3): 192-199, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-37407277

ABSTRACT

INTRODUCTION: Spinal cord injury (SCI) is one of the most dreaded complications after spinal cord stimulation (SCS) implantation surgery. As a result, intraoperative neurophysiological monitoring (IONM) has been proposed to avoid accidental damage to nervous structures under anesthesia and confirm positioning for optimal stimulation. Our study uses a large administrative claims database to determine the 30-day risk of SCI after SCS implantation. METHODS: This retrospective cohort study used the IBM MarketScan Commercial and Medicare Supplemental Databases from 2016 to 2019. Adult patients undergoing SCS surgical procedures with at least 90 days of follow-up, IONM use, the type of sedation used during the procedure, and subsequent SCI were identified using administrative codes. In addition, logistic regression was used to examine the relationship between various risk factors and subsequent SCI. RESULTS: A total of 9676 patients underwent SCS surgery (64.7% percutaneous implants) during the study period. Nine hundred and forty-four (9.75%) patients underwent SCS implantation with IONM. Conscious sedation, Monitored Anesthesia Care anesthesia, and general anesthesia were used in patients with 0.9%, 60.2%, and 28.6%, respectively. Eighty-one (0.8%) patients developed SCI within 30 days after SCS implant surgery. The SCI rate was higher in the group that underwent IONM (2% vs 0.7%, p value <0.001) during the implantation procedure, reflecting the underlying risk. After adjustment for other factors, the OR of SCI is 2.39 (95% CI: 1.33 to 4.14, p value=0.002) times higher for those with IONM than those without IONM. CONCLUSIONS: Increased SCI risk among patients with IONM likely reflects higher baseline risk, and further research is needed for risk mitigation.


Subject(s)
Intraoperative Neurophysiological Monitoring , Spinal Cord Injuries , Spinal Cord Stimulation , Adult , Humans , Aged , United States , Intraoperative Neurophysiological Monitoring/methods , Retrospective Studies , Medicare , Spinal Cord Injuries/diagnosis , Spinal Cord Injuries/epidemiology , Spinal Cord Injuries/etiology , Spinal Cord Stimulation/adverse effects , Spinal Cord Stimulation/methods , Anesthesia, General/adverse effects , Spinal Cord
9.
Pain Pract ; 24(4): 659-669, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38102862

ABSTRACT

INTRODUCTION: Spinal cord stimulation (SCS) is a well-established treatment option for chronic pain. Pain over the implantable pulse generator, or pocket pain, is an incompletely understood risk of SCS implantation which may limit the efficacy of treatment and patient quality of life. The goal of this narrative review is to analyze the literature to gain a more thorough understanding of the incidence and risk factors for the development of pocket pain to help guide treatment options and minimize its occurrence in the future. METHODS: A literature review was conducted investigating the development of pocket pain in patients with SCS for the management of a variety of pain conditions. RESULTS: In total, 305 articles were included in the original database search and 50 met the criteria for inclusion. The highest level of evidence for papers that specifically investigated pocket pain was level III. Four retrospective, observational analyses included pocket pain as a primary outcome. The remainder of the included studies listed pocket pain as an adverse event of SCS implantation. CONCLUSIONS: There is a relative dearth of primary literature that examines the incidence, characteristics, and health economic implications of pocket pain in patients with SCS. This highlights the need for large-scale, high-quality prospective or randomized controlled trials examining pocket pain. This may ultimately help prevent and reduce pocket pain leading to improved efficacy of treatment and greater patient quality of life.


Subject(s)
Chronic Pain , Spinal Cord Stimulation , Humans , Prospective Studies , Retrospective Studies , Quality of Life , Chronic Pain/etiology , Chronic Pain/therapy , Spinal Cord Stimulation/adverse effects , Spinal Cord , Treatment Outcome
10.
Pain Pract ; 24(4): 673-676, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38102861

ABSTRACT

BACKGROUND: One of the unique advances in neuromodulation for chronic pain has been spinal cord stimulators (SCS) and dorsal root ganglion stimulators (DRG-S). These devices have aided in conditions such as neuropathic pain, complex regional pain syndromes, failed back surgery, and peripheral neuropathies. With these benefits, however, complications from implantable stimulators have included lead fractures and migration. The authors reviewed a lead migration, kinking, and subsequent fracture event involving a patient with complex regional pain syndrome (CRPS) II, who was treated with a DRG-S. CASE PRESENTATION: The case report follows this patient, from their past medical history to assessment of appropriate qualifications for neuromodulation, to successful surgical placement, to follow-up care. The authors further monitored assessment of inefficacy of pain relief, and identification of lead migration and kinking through imaging. In the process of removal, due to lead stress, lead fracturing occurred. After lead removal, the leads were fully replaced, and the patient was followed up and experienced improved pain relief. CONCLUSION: The case report assesses probable mechanisms of lead fracture and considerations for physicians for future assessment and triage of neuromodulation efficacy.


Subject(s)
Chronic Pain , Complex Regional Pain Syndromes , Electric Stimulation Therapy , Neuralgia , Spinal Cord Stimulation , Humans , Ganglia, Spinal , Complex Regional Pain Syndromes/etiology , Complex Regional Pain Syndromes/therapy , Electric Stimulation Therapy/adverse effects , Electric Stimulation Therapy/methods , Chronic Pain/etiology , Chronic Pain/therapy , Neuralgia/therapy , Spinal Cord Stimulation/adverse effects , Spinal Cord Stimulation/methods
11.
Article in English | MEDLINE | ID: mdl-38083629

ABSTRACT

There is a lack of measures that provide insights into how spinal cord stimulation (SCS) modulates nociceptive function in patients with persistent spinal pain syndrome type 2 (PSPS-T2). Recently, we observed altered nociceptive detection thresholds (NDTs) in response to intra-epidermal electrical stimulation (IES) on the feet of PSPS-T2 patients when dorsal root ganglion stimulation was turned on. Furthermore, we observed altered NDTs and evoked potentials (EPs) in response to IES on the hands of PSPS-T2 patients. To explore whether EPs were obstructed by SCS artifacts, we applied IES twice to the hands of patients with SCS turned on (SCS-ON/ON group). To explore possible confounding effects of SCS outside the stimulated area, we repeated IES on the hands of these patients, once with SCS turned off and subsequently once with SCS turned on (SCS-OFF/ON group). The results demonstrated that EPs were not obstructed by SCS artifacts. Additionally, NDTs and EPs did not significantly change between measurements in the SCS-ON/ON and the SCS-OFF/ON groups. Therefore, the results suggested that possible confounding effects of SCS outside the nociceptive system did not interfere with the detection task performance. This work warrants further exploration of NDT-EP phenomena in response to IES at the painful feet of patients.Clinical Relevance-This work contributes to developing a clinical tool to explore psychophysical and neurophysiological biomarkers for observing modulating effects of SCS in patients with PSPS-T2.


Subject(s)
Pain , Spinal Cord Stimulation , Humans , Pain/etiology , Spinal Cord Stimulation/adverse effects , Spinal Cord Stimulation/methods , Pain Management/methods , Pain Measurement/methods , Spinal Cord/physiology
12.
Curr Pain Headache Rep ; 27(12): 837-842, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38010489

ABSTRACT

PURPOSE OF REVIEW: Spinal cord stimulation (SCS) has been used for decades to alleviate chronic pain. A growing body of literature suggests that the procedure is not without risks. Understanding the risks of SCS implantation can help treating physicians formulate individualized care plans that promote patient safety and minimize risks. RECENT FINDINGS: The overall complication rate associated with SCS has been reported to be 31.9 to 43%. The most common complication in the literature appears to be electrode migration. Other complications ranging in rates of occurrence include hematoma formation, infection, spinal cord injury, and cerebrospinal fluid (CSF) leak. Case reports of syrinx formation, foreign body reaction, and fibrosis have also been described. Our review shows that there are strategies available to reduce and prevent complications. In addition, close monitoring and early intervention may prevent some of the adverse neurological outcomes. Nevertheless, additional research regarding patient and procedural factors is necessary to improve the safety profile of this intervention.


Subject(s)
Chronic Pain , Spinal Cord Injuries , Spinal Cord Stimulation , Humans , Prostheses and Implants , Chronic Pain/therapy , Chronic Pain/etiology , Spinal Cord Injuries/etiology , Spinal Cord Stimulation/adverse effects , Spinal Cord
13.
Trials ; 24(1): 580, 2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37691092

ABSTRACT

BACKGROUND: Spinal cord stimulation (SCS) is a surgical technique used in patients with chronic intractable pain, and its effectiveness and safety have been validated by multiple studies. However, to maintain an optimal and steady long-term effect is still challenging. Here, we report a new management paradigm integrating smartphone application and remote programming. Chronic pain patients with SCS implants can monitor their pain status on the phone and change stimulation parameters accordingly. The PreMaSy study is a randomized controlled trial to evaluate the clinical effectiveness and safety of this precise management system. METHODS: Patients with chronic intractable pain will be screened for eligibility, and 82 participants are anticipated to be enrolled in this trial. After the electrode implantation, the stimulation effectiveness will be tested. Participants with a reduction of more than 50% in the visual analog scale (VAS) will receive implantation of an implantable pulse generator and randomized (1:1) into the experimental group or control group. All participants will be asked to take online follow-ups and complete assessments using a smartphone application. Daily pain characteristic assessments and monthly quality of life questionnaires are integrated into the App, and participants will be required to complete these assessments. The daily VAS for pain intensity will be monitored and a threshold will be set based on baseline VAS score. The interventional appointment will be scheduled once the threshold is reached. The primary outcome is the health condition and quality of life assessed by the five-level EuroQol five-dimensional questionnaire (EQ-5D-5L). Utility values of EQ-5D-5L will be assessed at baseline and 1, 3, and 6 months post-operative. DISCUSSION: The PreMaSy study aims to evaluate the effectiveness and safety of a novel App-based, patient-centered, self-assessment management system for chronic intractable pain. A randomized controlled trial is designed to test the non-inferiority of this precise management system compared to the monthly online follow-ups. It is also expected to yield valuable experiences regarding precision medicine. TRIAL REGISTRATION: ClinicalTrials.gov NCT05761392. Registered on March 07, 2023.


Subject(s)
Chronic Pain , Pain, Intractable , Spinal Cord Stimulation , Humans , Spinal Cord Stimulation/adverse effects , Chronic Pain/diagnosis , Chronic Pain/therapy , Quality of Life , Prostheses and Implants , Randomized Controlled Trials as Topic
14.
Neuromodulation ; 26(7): 1400-1411, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37589641

ABSTRACT

OBJECTIVE: This study aimed to assess the safety and effectiveness of a new charge-distributed multiphase stimulation paradigm during an extended spinal cord stimulation (SCS) trial. MATERIALS AND METHODS: This prospective, multicenter, randomized, single-blind, feasibility study included participants with chronic low back and/or leg pain and baseline numerical rating scale (NRS) for overall pain intensity ≥6. After a successful commercial SCS trial, participants were randomized to multiphase SCS therapy A (approximately 600-1500 Hz) or B (approximately 300-600 Hz), delivered via an investigational external pulse generator and existing leads during an 11-to-12-day testing period. Primary end points were mean NRS change from baseline to final in-office visit for each multiphase therapy and between therapies. Secondary end points included mean NRS change from end of commercial trial to final study visit and incidence of device-related adverse events (AEs). Additional measures included patient-reported outcomes collected at home through electronic watches and written diaries. Power usage was compared between multiphase and commercial therapies. RESULTS: A total of 122 participants initiated a commercial trial; 77 were randomized to a multiphase arm, and 65 completed the study. Reductions in mean NRS scores from baseline to final study visit were significant for multiphase therapy A and B (-4.3 and -4.7, respectively; both p < 0.0001). There was no statistically significant difference in mean NRS reduction or percent pain relief between multiphase therapies. In an additional analysis, 63.9% of participants reported greater pain relief with multiphase than with commercial SCS therapy in the at-home setting. On average, multiphase required less power than did commercial devices. One non-serious device-related AE was reported, and no infections occurred during the extended trial. CONCLUSIONS: Multiphase SCS effectively reduced pain in participants with chronic low back and/or leg pain during a trial, with no unanticipated device-related AEs reported. Future studies should evaluate long-term effectiveness of multiphase stimulation. CLINICAL TRIAL REGISTRATION: The Clinicaltrials.gov registration number for the study is NCT03594266.


Subject(s)
Chronic Pain , Spinal Cord Stimulation , Humans , Spinal Cord Stimulation/adverse effects , Spinal Cord Stimulation/methods , Chronic Pain/diagnosis , Chronic Pain/therapy , Leg , Prospective Studies , Single-Blind Method , Treatment Outcome , Spinal Cord
15.
Neuromodulation ; 26(7): 1412-1423, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37486284

ABSTRACT

OBJECTIVES: The aim of this prospective, single-blinded, dose-response study was to evaluate the safety and efficacy of a novel, paresthesia-free (subperception) spinal cord stimulation (SCS) waveform designed to target dorsal horn dendrites for the treatment of chronic neuropathic low back pain (LBP). MATERIALS AND METHODS: Twenty-seven participants with chronic neuropathic LBP were implanted with a commercial SCS system after a successful trial of SCS therapy. Devices were programmed to deliver the investigative waveform (100 Hz, 1000 µs, T9/T10 bipole) at descending stimulation perception threshold amplitudes (80%, 60%, 40%). Programs were evaluated at six, ten, and 14 weeks, after which participants selected their preferred program, with more follow-up at 26 weeks (primary outcomes). Participants were blinded to the nature of the programming. Pain score (visual analog scale [VAS]), Brief Pain Inventory (BPI), quality of life (EQ-5D-5L), and health status (36-Item Short Form [SF-36]) were measured at baseline and follow-ups. Responder rate, treatment satisfaction, clinician global impression of change, and adverse events (AEs) also were evaluated. RESULTS: Mean (± SD) baseline VAS was 72.5 ± 11.2 mm. At 26 weeks (n = 26), mean change from baseline in VAS was -51.7 mm (95% CI, -60.7 to -42.7; p < 0.001), with 76.9% of participants reporting ≥50% VAS reduction, and 46.2% reporting ≥80% VAS reduction. BPI, EQ-5D-5L, and SF-36 scores were all statistically significantly improved at 26 weeks (p < 0.001), and 100% of participants were satisfied with their treatment. There were no unanticipated AEs related to the study intervention, device, or procedures. CONCLUSIONS: This novel, paresthesia-free stimulation waveform may be a safe and effective option for patients with chronic neuropathic LBP eligible for SCS therapy and is deliverable by all current commercial SCS systems. CLINICAL TRIAL REGISTRATION: This study is registered on anzctr.org.au with identifier ACTRN12618000647235.


Subject(s)
Chronic Pain , Low Back Pain , Peripheral Nervous System Diseases , Spinal Cord Stimulation , Humans , Chronic Pain/diagnosis , Chronic Pain/therapy , Low Back Pain/diagnosis , Low Back Pain/therapy , Paresthesia/diagnosis , Paresthesia/therapy , Prospective Studies , Quality of Life , Spinal Cord , Spinal Cord Stimulation/adverse effects , Spinal Cord Stimulation/methods , Treatment Outcome
16.
World Neurosurg ; 178: e300-e306, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37473865

ABSTRACT

OBJECTIVE: Spinal cord stimulation (SCS) and dorsal root entry zone (DREZ) lesioning are important therapeutic options for intractable post-traumatic neuropathic pain (PNP). However, surgical choice is controversial due to the need to maximize pain relief and reduce complications. This study aims to retrospectively analyze the effect and complications of DREZ lesioning for patients with PNP who were unresponsive to SCS and provide a surgical reference. METHODS: Demographic data and surgical characteristics of patients with PNP who underwent DREZ lesioning after an unresponsive SCS were reviewed. Long-term outcomes including numeric rating scale, global impression of change, and long-term complications were assessed. Kaplan-Meier analysis was used to evaluate pain-free survival. RESULTS: Of 19 patients with PNP, 8 had brachial plexus injury (BPI), 7 had spinal cord injury, 2 had cauda equina injury, 1 had intercostal nerve injury, and 1 had lumbosacral plexus injury. All patients were unresponsive or had a recurrence of pain after SCS, with an average pain-relief rate of 9.3%. After DREZ lesioning, the mean numeric rating scale scores significantly decreased from 7.6 ± 1.5 to 1.8 ± 1.7, with an average pain-relief rate of 75.3%. Seven patients (36.8%) experienced worsened neurologic dysfunction at the last follow-up. Patients with BPI had a significantly better outcome than other pathologies (P < 0.001) after DREZ lesioning. CONCLUSIONS: DREZ lesioning is an effective alternative procedure to SCS for patients with PNP who have lost limb function. Particularly for those with BPI, DREZ lesioning has shown good efficacy and can be considered a preferred surgical option.


Subject(s)
Brachial Plexus , Neuralgia , Spinal Cord Stimulation , Humans , Spinal Nerve Roots/surgery , Retrospective Studies , Spinal Cord Stimulation/adverse effects , Neuralgia/etiology , Neuralgia/therapy , Brachial Plexus/surgery , Brachial Plexus/injuries , Spinal Cord
17.
Pain Pract ; 23(8): 933-941, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37409553

ABSTRACT

OBJECTIVES: Spinal cord stimulation (SCS) is conventionally placed at either cervical or thoracic spinal regions to treat chronic pain. However, for patients with multiarea pain, concomitant cervical and thoracic SCS (ctSCS) may be necessary to provide sufficient coverage. It remains unknown whether ctSCS is effective and safe. Thus, we aimed to survey the existing literature and assess the efficacy and safety of ctSCS. METHODS: A systematic review of the literature was performed according to the 2020 PRISMA guidelines to investigate pain, functional, and safety outcomes related to ctSCS. Articles between 1990 and 2022 available through PubMed, Web of Science, Scopus, and Cochrane Library databases were included if they assessed these outcomes in the context of ctSCS. Data extracted from articles included study type, number of ctSCS implantations, stimulation parameters, indications for implantation, complications, and frequency. The Newcastle-Ottawa scale was used to assess risk of bias. RESULTS: Three primary studies met our inclusion criteria. Overall, ctSCS was effective in providing analgesia. Pain severity was captured with patient-reported pain scales and changes in analgesic requirements. Various metrics were used to quantify quality of life and functional outcomes. Failed back surgery syndrome was the most common indication for ctSCS implantation. Implanted pulse generator pocket pain was the most common postoperative adverse event. CONCLUSIONS: Despite the limited evidence available, ctSCS seems to be effective and generally well tolerated. The dearth of relevant primary literature illustrates a knowledge gap, and future studies are needed to better clarify the efficacy and safety profile of this SCS variant.


Subject(s)
Chronic Pain , Spinal Cord Stimulation , Humans , Chronic Pain/drug therapy , Spinal Cord Stimulation/adverse effects , Quality of Life , Pain Management/adverse effects , Analgesics/therapeutic use , Spinal Cord , Treatment Outcome
18.
Clin Spine Surg ; 36(10): E430-E434, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37482636

ABSTRACT

STUDY DESIGN: Retrospective cohort study. OBJECTIVE: To determine risks associated with posterior lumbar arthrodesis after spinal cord stimulator (SCS) and intrathecal pain pump (IPP) insertion. SUMMARY OF BACKGROUND DATA: SCS and IPPs aid in the management of chronic back and radicular pain. Little is known regarding the risks of subsequent fusion with these devices in place. METHODS: The PearlDiver Mariner database was queried for spinal fusion between 2010 and 2020. Study groups were created for indwelling SCS or IPP and matched to a separate cohort without SCS or IPP. Subgroups were created for those who had their device removed and those who retained their device at the time of surgery. Complications up to 2 years postoperatively were reviewed and confounding variables were controlled using multivariable logistic regression. RESULTS: Four thousand five hundred three patients had an indwelling SCS/IPP and underwent posterior lumbar fusion. Compared with patients without history of an SCS/IPP, patients undergoing a lumbar fusion with an SCS/IPP which was removed or retained had higher rates of revision surgery at 1 year [7.3% vs. 5.0%, odds ratio (OR) =1.48, P <0.001] and 2 year (10.8% vs. 7.0%, OR =1.59, P <0.001). For all time intervals, there were higher odds of instrumentation failure (2 y: OR =1.65, P <0.001), and pulmonary complications (2 y: OR =1.18, P <0.001). At 2 years, there were higher odds of surgical site complications (OR 1.15, P =0.02) and urinary complications (OR=1.07, P =0.04). There were no differences in complications up to 2 years postoperatively in patients with an SCS/IPP who had their devices retained or removed ( P >0.05). CONCLUSIONS: Patients with a history of spinal cord stimulators and intrathecal pain pumps are at increased risk of mechanical complications and revision fusion compared with patients without these devices. Patients with an SCS or IPP have similar rates of complications during lumbar fusion whether the device is removed or retained.


Subject(s)
Spinal Cord Stimulation , Spinal Fusion , Humans , Retrospective Studies , Spinal Cord Stimulation/adverse effects , Lumbosacral Region , Pain/etiology , Spinal Cord/surgery , Spinal Fusion/adverse effects , Lumbar Vertebrae/surgery , Postoperative Complications/etiology
19.
Pain Pract ; 23(8): 886-891, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37381678

ABSTRACT

BACKGROUND: Spinal cord stimulation (SCS) is a minimally invasive therapy that is increasingly used to treat refractory neuropathic pain. Although this technique has a low incidence of serious long-term adverse sequelae, the risk of complications such as inadvertent dural puncture remains. OBJECTIVES: The goal of this article was to determine the impact of the contralateral oblique (CLO) fluoroscopic view incidence of postdural puncture headache (PDPH) during spinal cord stimulator implantation as compared to lateral fluoroscopic view. METHODS: This was a single academic institution retrospective analysis of electronic medical records spanning an approximate 20-year time period. Operative and postoperative notes were reviewed for details on dural puncture, including technique and spinal level of access, the development of a PDPH, and subsequent management. RESULTS: Over nearly two decades, a total of 1637 leads inserted resulted in 5 PDPH that were refractory to conservative measures but responded to epidural blood patch without long-term complications. The incidence of PDPH per lead insertion utilizing loss of resistance and lateral fluoroscopic guidance was 0.8% (4/489). However, adoption of CLO guidance was associated with a lower rate of PDPH at 0.08% (1/1148), p < 0.02. CONCLUSIONS: The incorporation of the CLO view to guide epidural needle placement can decrease the odds of a PDPH during percutaneous SCS procedures. This study further provides real-world data supporting the potential enhanced accuracy of epidural needle placement in order to avoid unintentional puncture or trauma to deeper spinal anatomic structures.


Subject(s)
Anesthesia, Epidural , Post-Dural Puncture Headache , Spinal Cord Stimulation , Humans , Post-Dural Puncture Headache/epidemiology , Post-Dural Puncture Headache/etiology , Post-Dural Puncture Headache/therapy , Retrospective Studies , Spinal Cord Stimulation/adverse effects , Incidence , Anesthesia, Epidural/adverse effects , Blood Patch, Epidural/methods
20.
J Opioid Manag ; 19(2): 171-178, 2023.
Article in English | MEDLINE | ID: mdl-37270425

ABSTRACT

BACKGROUND: Spinal cord stimulation (SCS) is an effective therapy for neuropathic pain. Outcomes of SCS may be influenced by peri-implant opioid management; however, to date, standard practices of opioid management in this scenario remain undefined and unreported. METHODS: A survey inquiring SCS management practices in the peri-implant period was sent to the members of the Spine Intervention Society and the American Society of Regional Anesthesia. The results of three questions pertaining to peri-implant opioid management are presented here. RESULTS: For each of the three questions examined, there were between 181 and 195 responses. Among respondents, 40 percent encouraged reduction of opioids prior to SCS trial, and 17 percent mandated reduction. After a SCS trial, 87 percent of respondents did not provide any additional opioids for periprocedure pain. After implant, the majority of respondents provided 1-7 days of opioids for post-operative pain. CONCLUSION: Based upon survey results and current literature, it is advisable to recommend or attempt opioid reduction before SCS and to not provide additional opioid for post-operative pain after trial lead insertion. Routine prescribing for the pain of the SCS implant beyond 7 days is not favored.


Subject(s)
Chronic Pain , Physicians , Spinal Cord Stimulation , Humans , Analgesics, Opioid/adverse effects , Spinal Cord Stimulation/adverse effects , Spinal Cord Stimulation/methods , Practice Patterns, Physicians' , Pain, Postoperative/diagnosis , Pain, Postoperative/drug therapy , Pain, Postoperative/prevention & control , Chronic Pain/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL