Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 119
1.
Neurotherapeutics ; 21(3): e00366, 2024 Apr.
Article En | MEDLINE | ID: mdl-38688105

Psychiatric disorders are among the leading contributors to global disease burden and disability. A significant portion of patients with psychiatric disorders remain treatment-refractory to best available therapy. With insights from the neurocircuitry of psychiatric disorders and extensive experience of neuromodulation with deep brain stimulation (DBS) in movement disorders, DBS is increasingly being considered to modulate the neural network in psychiatric disorders. Currently, obsessive-compulsive disorder (OCD) is the only U.S. FDA (United States Food and Drug Administration) approved DBS indication for psychiatric disorders. Medically refractory depression, addiction, and other psychiatric disorders are being explored for DBS neuromodulation. Studies evaluating DBS for psychiatric disorders are promising but lack larger, controlled studies. This paper presents a brief review and the current state of DBS and other neurosurgical neuromodulation therapies for OCD and other psychiatric disorders. We also present a brief review of MR-guided Focused Ultrasound (MRgFUS), a novel form of neurosurgical neuromodulation, which can target deep subcortical structures similar to DBS, but in a noninvasive fashion. Early experiences of neurosurgical neuromodulation therapies, including MRgFUS neuromodulation are encouraging in psychiatric disorders; however, they remain investigational. Currently, DBS and VNS are the only FDA approved neurosurgical neuromodulation options in properly selected cases of OCD and depression, respectively.


Deep Brain Stimulation , Mental Disorders , Humans , Deep Brain Stimulation/methods , Mental Disorders/therapy , Obsessive-Compulsive Disorder/therapy , Neurosurgical Procedures/methods , Neurosurgical Procedures/trends
2.
Proc Natl Acad Sci U S A ; 121(11): e2316365121, 2024 Mar 12.
Article En | MEDLINE | ID: mdl-38451949

Visceral signals are constantly processed by our central nervous system, enable homeostatic regulation, and influence perception, emotion, and cognition. While visceral processes at the cortical level have been extensively studied using non-invasive imaging techniques, very few studies have investigated how this information is processed at the single neuron level, both in humans and animals. Subcortical regions, relaying signals from peripheral interoceptors to cortical structures, are particularly understudied and how visceral information is processed in thalamic and subthalamic structures remains largely unknown. Here, we took advantage of intraoperative microelectrode recordings in patients undergoing surgery for deep brain stimulation (DBS) to investigate the activity of single neurons related to cardiac and respiratory functions in three subcortical regions: ventral intermedius nucleus (Vim) and ventral caudalis nucleus (Vc) of the thalamus, and subthalamic nucleus (STN). We report that the activity of a large portion of the recorded neurons (about 70%) was modulated by either the heartbeat, the cardiac inter-beat interval, or the respiration. These cardiac and respiratory response patterns varied largely across neurons both in terms of timing and their kind of modulation. A substantial proportion of these visceral neurons (30%) was responsive to more than one of the tested signals, underlining specialization and integration of cardiac and respiratory signals in STN and thalamic neurons. By extensively describing single unit activity related to cardiorespiratory function in thalamic and subthalamic neurons, our results highlight the major role of these subcortical regions in the processing of visceral signals.


Deep Brain Stimulation , Subthalamic Nucleus , Animals , Humans , Thalamus/physiology , Neurons/physiology , Microelectrodes
3.
N Engl J Med ; 390(1): 55-62, 2024 Jan 04.
Article En | MEDLINE | ID: mdl-38169490

Antiamyloid antibodies have been used to reduce cerebral amyloid-beta (Aß) load in patients with Alzheimer's disease. We applied focused ultrasound with each of six monthly aducanumab infusions to temporarily open the blood-brain barrier with the goal of enhancing amyloid removal in selected brain regions in three participants over a period of 6 months. The reduction in the level of Aß was numerically greater in regions treated with focused ultrasound than in the homologous regions in the contralateral hemisphere that were not treated with focused ultrasound, as measured by fluorine-18 florbetaben positron-emission tomography. Cognitive tests and safety evaluations were conducted over a period of 30 to 180 days after treatment. (Funded by the Harry T. Mangurian, Jr. Foundation and the West Virginia University Rockefeller Neuroscience Institute.).


Alzheimer Disease , Blood-Brain Barrier , Ultrasonic Therapy , Humans , Alzheimer Disease/diagnosis , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/analysis , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/diagnostic imaging , Positron-Emission Tomography/methods , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use
4.
J Neurosurg ; 140(1): 231-239, 2024 Jan 01.
Article En | MEDLINE | ID: mdl-37329519

OBJECTIVE: There were more than 107,000 drug overdose deaths in the US in 2021, the most ever recorded. Despite advances in behavioral and pharmacological treatments, over 50% of those receiving treatment for opioid use disorder (OUD) experience drug use recurrence (relapse). Given the prevalence of OUD and other substance use disorders (SUDs), the high rate of drug use recurrence, and the number of drug overdose deaths, novel treatment strategies are desperately needed. The objective of this study was to evaluate the safety and feasibility of deep brain stimulation (DBS) targeting the nucleus accumbens (NAc)/ventral capsule (VC) and potential impact on outcomes in individuals with treatment-refractory OUD. METHODS: A prospective, open-label, single-arm study was conducted among participants with longstanding treatment-refractory OUD (along with other co-occurring SUDs) who underwent DBS in the NAc/VC. The primary study endpoint was safety; secondary/exploratory outcomes included opioid and other substance use, substance craving, and emotional symptoms throughout follow-up and 18FDG-PET neuroimaging. RESULTS: Four male participants were enrolled and all tolerated DBS surgery well with no serious adverse events (AEs) and no device- or stimulation-related AEs. Two participants sustained complete substance abstinence for > 1150 and > 520 days, respectively, with significant post-DBS reductions in substance craving, anxiety, and depression. One participant experienced post-DBS drug use recurrences with reduced frequency and severity. The DBS system was explanted in one participant due to noncompliance with treatment requirements and the study protocol. 18FDG-PET neuroimaging revealed increased glucose metabolism in the frontal regions for the participants with sustained abstinence only. CONCLUSIONS: DBS of the NAc/VC was safe, feasible, and can potentially reduce substance use, craving, and emotional symptoms in those with treatment-refractory OUD. A randomized, sham-controlled trial in a larger cohort of patients is being initiated.


Deep Brain Stimulation , Drug Overdose , Opioid-Related Disorders , Humans , Male , Nucleus Accumbens/diagnostic imaging , Deep Brain Stimulation/methods , Fluorodeoxyglucose F18 , Prospective Studies , Feasibility Studies , Neoplasm Recurrence, Local , Opioid-Related Disorders/therapy
5.
Front Psychiatry ; 14: 1211566, 2023.
Article En | MEDLINE | ID: mdl-37779628

Introduction: While current treatments for substance use disorder (SUD) are beneficial, success rates remain low and treatment outcomes are complicated by co-occurring SUDs, many of which are without available medication treatments. Research involving neuromodulation for SUD has recently gained momentum. This study evaluated two doses (60 and 90 W) of Low Intensity Focused Ultrasound (LIFU), targeting the bilateral nucleus accumbens (NAc), in individuals with SUD. Methods: Four participants (three male), who were receiving comprehensive outpatient treatment for opioid use disorder at the time of enrollment and who also had a history of excessive non-opioid substance use, completed this pilot study. After confirming eligibility, these participants received 10 min sham LIFU followed by 20 min active LIFU (10 min to left then right NAc). Outcomes were the safety, tolerability, and feasibility during the LIFU procedure and throughout the 90-day follow-up. Outcomes also included the impact of LIFU on cue-induced substance craving, assessed via Visual Analog Scale (VAS), both acutely (pre-, during and post-procedure) and during the 90-day follow-up. Daily craving ratings (without cues) were also obtained for one-week prior to and one-week following LIFU. Results: Both LIFU doses were safe and well-tolerated based on reported adverse events and MRI scans revealed no structural changes (0 min, 24 h, and 1-week post-procedure). For the two participants receiving "enhanced" (90 W) LIFU, VAS craving ratings revealed active LIFU attenuated craving for participants' primary substances of choice relative to sham sonication. For these participants, reductions were also noted in daily VAS craving ratings (0 = no craving; 10 = most craving ever) across the week following LIFU relative to pre-LIFU; Participant #3 pre- vs. post-LIFU: opioids (3.6 ± 0.6 vs. 1.9 ± 0.4), heroin (4.2 ± 0.8 vs. 1.9 ± 0.4), methamphetamine (3.2 ± 0.4 vs. 0.0 ± 0.0), cocaine (2.4 ± 0.6 vs. 0.0 ± 0.0), benzodiazepines (2.8 ± 0.5 vs. 0.0 ± 0.0), alcohol (6.0 ± 0.7 vs. 2.7 ± 0.8), and nicotine (5.6 ± 1.5 vs. 3.1 ± 0.7); Participant #4: alcohol (3.5 ± 1.3 vs. 0.0 ± 0.0) and nicotine (5.0 ± 1.8 vs. 1.2 ± 0.8) (all p's < 0.05). Furthermore, relative to screening, longitudinal reductions in cue-induced craving for several substances persisted during the 90-day post-LIFU follow-up evaluation for all participants. Discussion: In conclusion, LIFU targeting the NAc was safe and acutely reduced substance craving during the LIFU procedure, and potentially had longer-term impact on craving reductions. While early observations are promising, NAc LIFU requires further investigation in a controlled trial to assess the impact on substance craving and ultimately substance use and relapse.

7.
Drug Alcohol Depend ; 249: 110817, 2023 Aug 01.
Article En | MEDLINE | ID: mdl-37331302

BACKGROUND: Identifying predictors of drug use recurrence (DUR) is critical to combat the addiction epidemic. Wearable devices and phone-based applications for obtaining self-reported assessments in the patient's natural environment (e.g., ecological momentary assessment; EMA) have been used in various healthcare settings. However, the utility of combining these technologies to predict DUR in substance use disorder (SUD) has not yet been explored. This study investigates the combined use of wearable technologies and EMA as a potential mechanism for identifying physiological/behavioral biomarkers of DUR. METHODS: Participants, recruited from an SUD treatment program, were provided with a commercially available wearable device that continuously monitors biometric signals (e.g., heart rate/variability [HR/HRV], sleep characteristics). They were also prompted daily to complete an EMA via phone-based application (EMA-APP) that included questionnaires regarding mood, pain, and craving. RESULTS: Seventy-seven participants are included in this pilot study (34 participants experienced a DUR during enrollment). Wearable technologies revealed that physiological markers were significantly elevated in the week prior to DUR relative to periods of sustained abstinence (p<0.001). Results from the EMA-APP revealed that those who experienced a DUR reported greater difficulty concentrating, exposure to triggers associated with substance use, and increased isolation the day prior to DUR (p<0.001). Compliance with study procedures during the DUR week was lower than any other period of measurement (p<0.001). CONCLUSIONS: These results suggest that data acquired via wearable technologies and the EMA-APP may serve as a method of predicting near-term DUR, thereby potentially prompting intervention before drug use occurs.


Substance-Related Disorders , Wearable Electronic Devices , Humans , Pilot Projects , Substance-Related Disorders/diagnosis , Surveys and Questionnaires , Smartphone , Ecological Momentary Assessment
8.
Fluids Barriers CNS ; 20(1): 46, 2023 Jun 16.
Article En | MEDLINE | ID: mdl-37328855

BACKGROUND: Focused ultrasound (FUS)-mediated blood-brain barrier (BBB) opening is under investigation as a therapeutic modality for neurodegeneration, yet its effects in humans are incompletely understood. Here, we assessed physiologic responses to FUS administered in multifocal brain sites of persons with Alzheimer's disease (AD). METHODS: At a tertiary neuroscience institute, eight participants with AD (mean age 65, 38% F) enrolled in a phase 2 clinical trial underwent three successive targeted BBB opening procedures at 2 week intervals using a 220 kHz FUS transducer in combination with systemically administered microbubbles. In all, 77 treatment sites were evaluated and encompassed hippocampal, frontal, and parietal brain regions. Post-FUS imaging changes, including susceptibility effects and spatiotemporal gadolinium-based contrast agent enhancement patterns, were analyzed using serial 3.0-Tesla MRI. RESULTS: Post-FUS MRI revealed expected intraparenchymal contrast extravasation due to BBB opening at all targeted brain sites. Immediately upon BBB opening, hyperconcentration of intravenously-administered contrast tracer was consistently observed around intracerebral veins. Following BBB closure, within 24-48 h of FUS intervention, permeabilization of intraparenchymal veins was observed and persisted for up to one week. Notably, extraparenchymal meningeal venous permeabilization and associated CSF effusions were also elicited and persisted up to 11 days post FUS treatment, prior to complete spontaneous resolution in all participants. Mild susceptibility effects were detected, however no overt intracranial hemorrhage or other serious adverse effects occurred in any participant. CONCLUSIONS: FUS-mediated BBB opening is safely and reproducibly achieved in multifocal brain regions of persons with AD. Post-FUS tracer enhancement phenomena suggest the existence of a brain-wide perivenous fluid efflux pathway in humans and demonstrate reactive physiological changes involving these conduit spaces in the delayed, subacute phase following BBB disruption. The delayed reactive venous and perivenous changes are consistent with a dynamic, zonal exudative response to upstream capillary manipulation. Further preclinical and clinical investigations of these FUS-related imaging phenomena and of intracerebral perivenous compartment changes are needed to elucidate physiology of this pathway as well as biological effects of FUS administered with and without adjuvant neurotherapeutics. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT03671889, registered 9/14/2018.


Alzheimer Disease , Blood-Brain Barrier , Aged , Humans , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Ultrasonography , Male , Female
9.
Pediatr Neurol ; 142: 47-50, 2023 05.
Article En | MEDLINE | ID: mdl-36907087

The AIFM1 gene encodes a mitochondrial protein that acts as a flavin adenine dinucleotide-dependent nicotinamide adenine dinucleotide oxidase and apoptosis regulator. Monoallelic pathogenic AIFM1 variants result in a spectrum of X-linked neurological disorders, including Cowchock syndrome. Common features in Cowchock syndrome include a slowly progressive movement disorder, cerebellar ataxia, progressive sensorineural hearing loss, and sensory neuropathy. We identified a novel maternally inherited hemizygous missense AIFM1 variant, c.1369C>T p.(His457Tyr), in two brothers with clinical features consistent with Cowchock syndrome using next-generation sequencing. Both individuals had a progressive complex movement disorder phenotype, including disabling tremor poorly responsive to medications. Deep brain stimulation (DBS) of the ventral intermediate thalamic nucleus ameliorated contralateral tremor and improved their quality of life; this suggests the beneficial role for DBS in treatment-resistant tremor within AIFM1-related disorders.


Charcot-Marie-Tooth Disease , Deep Brain Stimulation , Humans , Male , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Quality of Life , Tremor/genetics , Tremor/therapy
10.
J Neurosurg ; 139(1): 275-283, 2023 07 01.
Article En | MEDLINE | ID: mdl-36334289

OBJECTIVE: MRI-guided low-intensity focused ultrasound (FUS) has been shown to reversibly open the blood-brain barrier (BBB), with the potential to deliver therapeutic agents noninvasively to target brain regions in patients with Alzheimer's disease (AD) and other neurodegenerative conditions. Previously, the authors reported the short-term safety and feasibility of FUS BBB opening of the hippocampus and entorhinal cortex (EC) in patients with AD. Given the need to treat larger brain regions beyond the hippocampus and EC, brain volumes and locations treated with FUS have now expanded. To evaluate any potential adverse consequences of BBB opening on disease progression, the authors report safety, imaging, and clinical outcomes among participants with mild AD at 6-12 months after FUS treatment targeted to the hippocampus, frontal lobe, and parietal lobe. METHODS: In this open-label trial, participants with mild AD underwent MRI-guided FUS sonication to open the BBB in ß-amyloid positive regions of the hippocampus, EC, frontal lobe, and parietal lobe. Participants underwent 3 separate FUS treatment sessions performed 2 weeks apart. Outcome assessments included safety, imaging, neurological, cognitive, and florbetaben ß-amyloid PET. RESULTS: Ten participants (range 55-76 years old) completed 30 separate FUS treatments at 2 participating institutions, with 6-12 months of follow-up. All participants had immediate BBB opening after FUS and BBB closure within 24-48 hours. All FUS treatments were well tolerated, with no serious adverse events related to the procedure. All 10 participants had a minimum of 6 months of follow-up, and 7 participants had a follow-up out to 1 year. Changes in the Alzheimer's Disease Assessment Scale-cognitive and Mini-Mental State Examination scores were comparable to those in controls from the Alzheimer's Disease Neuroimaging Initiative. PET scans demonstrated an average ß-amyloid plaque of 14% in the Centiloid scale in the FUS-treated regions. CONCLUSIONS: This study is the largest cohort of participants with mild AD who received FUS treatment, and has the longest follow-up to date. Safety was demonstrated in conjunction with reversible and repeated BBB opening in multiple cortical and deep brain locations, with a concomitant reduction of ß-amyloid. There was no apparent cognitive worsening beyond expectations up to 1 year after FUS treatment, suggesting that the BBB opening treatment in multiple brain regions did not adversely influence AD progression. Further studies are needed to determine the clinical significance of these findings. FUS offers a unique opportunity to decrease amyloid plaque burden as well as the potential to deliver targeted therapeutics to multiple brain regions in patients with neurodegenerative disorders.


Alzheimer Disease , Blood-Brain Barrier , Humans , Middle Aged , Aged , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/metabolism , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/therapy , Plaque, Amyloid , Brain/metabolism , Amyloid beta-Peptides/metabolism , Cognition
11.
Article En | MEDLINE | ID: mdl-36119969

Background: High frequency focused ultrasound is used for treatment of essential tremor. Side effects associated with the procedure may resolve over time. We report a case of negative myoclonus, which has not been reported with this procedure. Case report: A 73-year-old left-handed man underwent focused ultrasound thalamotomy for treatment of essential tremor. Immediately post procedure he was noted to have negative myoclonus in the treated limb. This side effect resolved over the course of 6 months. Discussion: Although asterixis has been associated with thalamic infarcts in the past, this has not yet been reported in the literature with MRgFUS procedure and is a novel observation. Occupational and physical therapy may be considered to address this side effect. It is important to counsel patients about the rare occurrence of this complication of therapy but also its potential for complete resolution over time.


Deep Brain Stimulation , Essential Tremor , Myoclonus , Aged , Deep Brain Stimulation/methods , Essential Tremor/diagnostic imaging , Essential Tremor/surgery , Humans , Male , Myoclonus/therapy , Thalamus/diagnostic imaging , Thalamus/surgery , Ultrasonography/methods
12.
J Neurol Sci ; 437: 120253, 2022 06 15.
Article En | MEDLINE | ID: mdl-35460949

While pharmacological and/or behavioral treatments are effective in managing symptoms for many patients with psychiatric diagnoses and disorders with behavioral/cognitive manifestations, a subset of individuals are treatment-refractory, unable to achieve appreciable benefit or symptom relief from traditional methods. In recent years, neuromodulation has gained momentum as an adjunctive treatment for improving outcomes in patients who are treatment-refractory. One form of neuromodulation, deep brain stimulation (DBS), has been investigated for the treatment of various psychiatric disorders and behavioral/cognitive symptoms. The following article provides a review of DBS investigations for several psychiatric and behavioral-related disorders, including depression, obsessive-compulsive disorder, substance use disorder, Alzheimer's disease, anorexia, obesity, schizophrenia, and posttraumatic stress disorder. PubMed, PsycINFO, Scopus, Ovid MEDLINE, and Web of Science were used to identify published articles, and Clinicaltrials.gov was used to identify currently ongoing or planned studies. Findings revealed the potential utility of DBS in improving outcomes for various psychiatric and behavioral/cognitive-related disorders. While promising, there are several limitations present in the available literature, and further well-designed clinical trials are necessary before conclusive decisions regarding the utility of DBS for the treatment of these psychiatric/behavioral/cognitive-related disorders can be made. Regardless, the studies included in this review demonstrate positive preliminary findings for the potential benefit of DBS for treatment of a variety of psychiatric disorders, and further research is warranted to better determine the potential utility of DBS for those who are treatment-refractory and unable to achieve symptom relief with standard care.


Deep Brain Stimulation , Obsessive-Compulsive Disorder , Stress Disorders, Post-Traumatic , Substance-Related Disorders , Cognition , Deep Brain Stimulation/methods , Humans
13.
PLoS One ; 16(10): e0257997, 2021.
Article En | MEDLINE | ID: mdl-34648513

Conventional testing and diagnostic methods for infections like SARS-CoV-2 have limitations for population health management and public policy. We hypothesize that daily changes in autonomic activity, measured through off-the-shelf technologies together with app-based cognitive assessments, may be used to forecast the onset of symptoms consistent with a viral illness. We describe our strategy using an AI model that can predict, with 82% accuracy (negative predictive value 97%, specificity 83%, sensitivity 79%, precision 34%), the likelihood of developing symptoms consistent with a viral infection three days before symptom onset. The model correctly predicts, almost all of the time (97%), individuals who will not develop viral-like illness symptoms in the next three days. Conversely, the model correctly predicts as positive 34% of the time, individuals who will develop viral-like illness symptoms in the next three days. This model uses a conservative framework, warning potentially pre-symptomatic individuals to socially isolate while minimizing warnings to individuals with a low likelihood of developing viral-like symptoms in the next three days. To our knowledge, this is the first study using wearables and apps with machine learning to predict the occurrence of viral illness-like symptoms. The demonstrated approach to forecasting the onset of viral illness-like symptoms offers a novel, digital decision-making tool for public health safety by potentially limiting viral transmission.


Artificial Intelligence , COVID-19/diagnosis , Health Personnel , Models, Theoretical , Wearable Electronic Devices , Humans , Machine Learning , Pilot Projects , Sensitivity and Specificity
14.
Exp Clin Psychopharmacol ; 29(2): 210-215, 2021 Apr.
Article En | MEDLINE | ID: mdl-34043402

Given high relapse rates and the prevalence of overdose deaths, novel treatments for substance use disorder (SUD) are desperately needed for those who are treatment refractory. The objective of this study was to evaluate the safety of deep brain stimulation (DBS) for SUD and the effects of DBS on substance use, substance craving, emotional symptoms, and frontal/executive functions. DBS electrodes were implanted bilaterally within the Nucleus Accumbens/Ventral anterior internal capsule (NAc/VC) of a man in his early 30s with >10-year history of severe treatment refractory opioid and benzodiazepine use disorders. DBS of the NAc/VC was found to be safe with no serious adverse events noted and the participant remained abstinent and engaged in comprehensive treatment at the 12-week endpoint (and 12-month extended follow-up). Using a 0-100 visual analog scale, substance cravings decreased post-DBS implantation; most substantially in benzodiazepine craving following the final DBS titration (1.0 ± 2.2) compared to baseline (53.4 ± 29.5; p < .001). A trend toward improvement in frontal/executive function was observed on the balloon analog risk task performance following the final titration (217.7 ± 76.2) compared to baseline (131.3 ± 28.1, p = .066). FDG PET demonstrated an increase in glucose metabolism in the dorsolateral prefrontal and medial premotor cortices at the 12-week endpoint compared to post-surgery/pre-DBS titration. Heart Rate Variability (HRV) improved following the final titration (rMSSD = 56.0 ± 11.7) compared to baseline (19.2 ± 8.2; p < .001). In a participant with severe, treatment refractory opioid and benzodiazepine use disorder, DBS of the NAc/VC was safe, reduced substance use and craving, and improved frontal and executive functions. Confirmation of these findings with future studies is needed. (PsycInfo Database Record (c) 2021 APA, all rights reserved).


Benzodiazepines , Deep Brain Stimulation , Nucleus Accumbens , Substance-Related Disorders/therapy , Adult , Analgesics, Opioid/adverse effects , Benzodiazepines/adverse effects , Humans , Internal Capsule , Male , Pilot Projects
15.
Front Sports Act Living ; 3: 585870, 2021.
Article En | MEDLINE | ID: mdl-33733234

Commercial off-the shelf (COTS) wearable devices continue development at unprecedented rates. An unfortunate consequence of their rapid commercialization is the lack of independent, third-party accuracy verification for reported physiological metrics of interest, such as heart rate (HR) and heart rate variability (HRV). To address these shortcomings, the present study examined the accuracy of seven COTS devices in assessing resting-state HR and root mean square of successive differences (rMSSD). Five healthy young adults generated 148 total trials, each of which compared COTS devices against a validation standard, multi-lead electrocardiogram (mECG). All devices accurately reported mean HR, according to absolute percent error summary statistics, although the highest mean absolute percent error (MAPE) was observed for CameraHRV (17.26%). The next highest MAPE for HR was nearly 15% less (HRV4Training, 2.34%). When measuring rMSSD, MAPE was again the highest for CameraHRV [112.36%, concordance correlation coefficient (CCC): 0.04], while the lowest MAPEs observed were from HRV4Training (4.10%; CCC: 0.98) and OURA (6.84%; CCC: 0.91). Our findings support extant literature that exposes varying degrees of veracity among COTS devices. To thoroughly address questionable claims from manufacturers, elucidate the accuracy of data parameters, and maximize the real-world applicative value of emerging devices, future research must continually evaluate COTS devices.

17.
Radiology ; 298(3): 654-662, 2021 03.
Article En | MEDLINE | ID: mdl-33399511

Background Opening of the blood-brain barrier (BBB) induced with MRI-guided focused ultrasound has been shown in experimental animal models to reduce amyloid-ß plaque burden, improve memory performance, and facilitate delivery of therapeutic agents to the brain. However, physiologic effects of this procedure in humans with Alzheimer disease (AD) require further investigation. Purpose To assess imaging effects of focused ultrasound-induced BBB opening in the hippocampus of human participants with early AD and to evaluate fluid flow patterns after BBB opening by using serial contrast-enhanced MRI. Materials and Methods Study participants with early AD recruited to a Health Insurance Portability and Accountability Act-compliant, prospective, ongoing phase II clinical trial (ClinicalTrials.gov identifier, NCT03671889) underwent three separate focused ultrasound-induced BBB opening procedures that used a 220-kHz transducer with a concomitant intravenous microbubble contrast agent administered at 2-week intervals targeting the hippocampus and entorhinal cortex between October 2018 and May 2019. Posttreatment effects and gadolinium-based contrast agent enhancement patterns were evaluated by using 3.0-T MRI. Results Three women (aged 61, 72, and 73 years) consecutively enrolled in the trial successfully completed repeated focused ultrasound-induced BBB opening of the hippocampus and entorhinal cortex. Postprocedure contrast enhancement was clearly identified within the targeted brain volumes, indicating immediate spatially precise BBB opening. Parenchymal enhancement resolved within 24 hours after all treatments, confirming BBB closure. Transient perivenous enhancement was consistently observed during the acute phase after BBB opening. Notably, contrast enhancement reappeared in the perivenular regions after BBB closure. This imaging marker is consistent with blood-meningeal barrier permeability and persisted for 24-48 hours before spontaneous resolution. No evidence of intracranial hemorrhage or other adverse effect was identified. Conclusion MRI-guided focused ultrasound-induced blood-brain barrier opening was safely performed in the hippocampi of three participants with Alzheimer disease without any adverse effects. Posttreatment MRI reveals a unique spatiotemporal contrast enhancement pattern that suggests a perivenular immunologic healing response downstream from targeted sites. © RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Klibanov in this issue.


Alzheimer Disease/diagnostic imaging , Alzheimer Disease/drug therapy , Blood-Brain Barrier/diagnostic imaging , Drug Delivery Systems/methods , Magnetic Resonance Imaging, Interventional/methods , Ultrasonic Therapy/methods , Aged , Contrast Media , Entorhinal Cortex , Female , Hippocampus , Humans , Middle Aged , Prospective Studies
18.
Neurol India ; 68(Supplement): S218-S223, 2020.
Article En | MEDLINE | ID: mdl-33318354

BACKGROUND: Spinal cord stimulation (SCS) has emerged as state-of-the-art evidence-based treatment for chronic intractable pain related to spinal and peripheral nerve disorders. Traditionally delivered as steady-state, paraesthesia-producing electrical stimulation, newer technology has augmented the SCS option and outcome in the last decade. OBJECTIVE: To present an overview of the traditional and newer SCS waveforms. MATERIALS AND METHODS: We present a short literature review of SCS waveforms in reference to newer waveforms and describing paraesthesia-free, high frequency, and burst stimulation methods as well as advances in waveform paradigms and programming modalities. Pertinent literature was reviewed, especially in the context of evolution in the waveforms of SCS and stimulation parameters. RESULTS: Conventional tonic SCS remains one of the most utilized and clinically validated SCS waveforms. Newer waveforms such as burst stimulation, high-frequency stimulation, and the sub-perception SCS have emerged in the last decades with favorable results with no or minimal paraesthesia, including in cases otherwise intractable to conventional tonic SCS. The recent evolution and experience of closed-loop SCS is promising and appealing. The experience and validation of the newer SCS waveforms, however, remain limited but optimistic. CONCLUSIONS: Advances in SCS device technology and waveforms have improved patient outcomes, leading to its increased utilization of SCS for chronic pain. These improvements and the development of closed-loop SCS have been increasingly promising development and foster a clinical translation of improved pain relief as the years of research and clinical study beyond conventional SCS waveform come to fruition.


Chronic Pain , Spinal Cord Stimulation , Chronic Pain/therapy , Humans , Pain Management
19.
Front Hum Neurosci ; 14: 593672, 2020.
Article En | MEDLINE | ID: mdl-33132889

The blood-brain barrier (BBB) limits therapeutic delivery in Alzheimer's disease (AD) and other neurological disorders. Animal models have demonstrated safe BBB opening and reduction in ß-amyloid plaque with focused ultrasound (FUS). We recently demonstrated the feasibility, safety, and reversibility of FUS-induced BBB opening in the hippocampus and entorhinal cortex in six participants with early AD. We now report the effect of BBB opening with FUS treatment on ß-amyloid plaque. Six participants underwent 18F-Florbetaben PET scan at baseline and 1 week after the completion of the third FUS treatment (60 days interval). PET analysis comparing the hippocampus and entorhinal cortex in the treated and untreated hemispheres revealed a decrease in the ratio of 18F-Florbetaben ligand binding. The standard uptake value ratios (SUVr) reduction ranged from 2.7% to 10% with an average of 5.05% (±2.76) suggesting a decrease in ß-amyloid plaque.

20.
J Neurol Sci ; 418: 117149, 2020 11 15.
Article En | MEDLINE | ID: mdl-33002757

Given the high prevalence of individuals diagnosed with substance use disorder, along with the elevated rate of relapse following treatment initiation, investigating novel approaches and new modalities for substance use disorder treatment is of vital importance. One such approach involves neuromodulation which has been used therapeutically for neurological and psychiatric disorders and has demonstrated positive preliminary findings for the treatment of substance use disorder. The following article provides a review of several forms of neuromodulation which warrant consideration as potential treatments for substance use disorder. PubMed, PsycINFO, Ovid MEDLINE, and Web of Science were used to identify published articles and clinicaltrials.gov was used to identify currently ongoing or planned studies. Search criteria for Brain Stimulation included the following terminology: transcranial direct current stimulation, transcranial magnetic stimulation, theta burst stimulation, deep brain stimulation, vagus nerve stimulation, trigeminal nerve stimulation, percutaneous nerve field stimulation, auricular nerve stimulation, and low intensity focused ultrasound. Search criteria for Addiction included the following terminology: addiction, substance use disorder, substance-related disorder, cocaine, methamphetamine, amphetamine, alcohol, nicotine, tobacco, smoking, marijuana, cannabis, heroin, opiates, opioids, and hallucinogens. Results revealed that there are currently several forms of neuromodulation, both invasive and non-invasive, which are being investigated for the treatment of substance use disorder. Preliminary findings have demonstrated the potential of these various neuromodulation techniques in improving substance treatment outcomes by reducing those risk factors (e.g. substance craving) associated with relapse. Specifically, transcranial magnetic stimulation has shown the most promise with several well-designed studies supporting the potential for reducing substance craving. Deep brain stimulation has also shown promise, though lacks well-controlled clinical trials to support its efficacy. Transcranial direct current stimulation has also demonstrated promising results though consistently designed, randomized trials are also needed. There are several other forms of neuromodulation which have not yet been investigated clinically but warrant further investigation given their mechanisms and potential efficacy based on findings from other studied indications. In summary, given promising findings in reducing substance use and craving, neuromodulation may provide a non-pharmacological option as a potential treatment and/or treatment augmentation for substance use disorder. Further research investigating neuromodulation, both alone and in combination with already established substance use disorder treatment (e.g. medication treatment), warrants consideration.


Deep Brain Stimulation , Substance-Related Disorders , Transcranial Direct Current Stimulation , Vagus Nerve Stimulation , Humans , Substance-Related Disorders/therapy , Transcranial Magnetic Stimulation
...