Skip to main content
Erschienen in: Journal of Neurology 11/2023

Open Access 02.08.2023 | Review

Machine learning for adaptive deep brain stimulation in Parkinson’s disease: closing the loop

verfasst von: Andreia M. Oliveira, Luis Coelho, Eduardo Carvalho, Manuel J. Ferreira-Pinto, Rui Vaz, Paulo Aguiar

Erschienen in: Journal of Neurology | Ausgabe 11/2023

Abstract

Parkinson’s disease (PD) is the second most common neurodegenerative disease bearing a severe social and economic impact. So far, there is no known disease modifying therapy and the current available treatments are symptom oriented. Deep Brain Stimulation (DBS) is established as an effective treatment for PD, however current systems lag behind today’s technological potential. Adaptive DBS, where stimulation parameters depend on the patient’s physiological state, emerges as an important step towards “smart” DBS, a strategy that enables adaptive stimulation and personalized therapy. This new strategy is facilitated by currently available neurotechnologies allowing the simultaneous monitoring of multiple signals, providing relevant physiological information. Advanced computational models and analytical methods are an important tool to explore the richness of the available data and identify signal properties to close the loop in DBS. To tackle this challenge, machine learning (ML) methods applied to DBS have gained popularity due to their ability to make good predictions in the presence of multiple variables and subtle patterns. ML based approaches are being explored at different fronts such as the identification of electrophysiological biomarkers and the development of personalized control systems, leading to effective symptom relief. In this review, we explore how ML can help overcome the challenges in the development of closed-loop DBS, particularly its role in the search for effective electrophysiology biomarkers. Promising results demonstrate ML potential for supporting a new generation of adaptive DBS, with better management of stimulation delivery, resulting in more efficient and patient-tailored treatments.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s00415-023-11873-1.

Introduction

Deep brain stimulation (DBS) is a therapy applied in an increasing range of neurological disorders, including Parkinson’s disease (PD). Second to Alzheimer’s disease, PD is one of the most common neurodegenerative diseases worldwide, with tremendous impact in society [1]. Currently available pharmacological treatments for PD are symptom oriented and there are no disease modifying therapies. These treatments achieve symptomatic control in a considerable proportion of patients but there is still a significant number of patients for whom pharmacologic therapies are ineffective or insufficient, leaving DBS therapy as the remaining option.
For the implementation of DBS in PD patients, an electrode is surgically placed in the brain, providing electrical stimulation patterns to a specific target area (the subthalamic nucleus (STN) or the globus pallidus pars interna (GPi)). In the initial setup after surgery, stimulation parameters are explored by the clinician in search for the configuration achieving the proper symptomatic control. Each parameter is tuned within a range of values where the intervention is potentially beneficial to the patient (for a brief overview of the tuneable parameters and their known effects see Table S1). Outside these subjective and patient dependent window’s boundaries, the therapy is not as effective and adverse effects might occur. Information on the precise lead location, obtained via imaging techniques, is also often taken into account in this process. The stimulation parameters are adjusted by a trial-and-error approach, relying on qualitative evaluation of clinical responses [2, 3]. The parameters are then kept fixed for the time between consultations. Given the dynamic nature of PD symptoms, this canonical open-loop stimulation strategy is suboptimal, since the applied therapy does not automatically adapt to the patient’s state nor condition evolution. Furthermore, its adverse effects (generally caused by the continuous stimulation with constant amplitude) are difficult to manage.
Progress towards optimized DBS (on both open and closed loop approaches) is challenged by the fact that there is still no clear understanding of its mechanisms of action on neuronal populations and circuits [4, 5]. While the exogenous electrical current applied by the electrode exerts direct effects on locally-residing cell bodies, which can be of inhibitory or excitatory nature, it also perturbs en passant axons, resulting in the modulation of complex neural networks [69]. Indeed, multiple mechanisms of action are likely to occur simultaneously. On one hand, this adds complexity to our understanding of the modulation process but, on the other hand, it also opens alternative potential targets to explore as to improve the neuromodulation results. Possible therapeutic mechanisms involve the direct suppression of local pathological neuronal activity, masking of pathological activity, and interactions with re-entrant nonlinear oscillations [7, 1015].
Adaptive DBS (aDBS) emerges as a promising paradigm to achieve “smart” DBS systems (Fig. 1). The principle behind this proposed approach is the optimization of stimulation parameters in direct response to the patient’s electrophysiological state, achievable in a closed-loop configuration. Closed-loop control mechanisms are associated with algorithms capable of automatically regulating the system’s parameters in order to maintain a desired/predefined target state, without human interaction. For aDBS, this means that DBS stimulation parameters continuously adjust to the patient’s varying needs. Considering the different possible strategies to close the loop in aDBS, the feedback systems directly based in neuronal signals are of particular interest. These feedback control systems become possible with real-time neuronal activity recordings obtained directly from the target region using dual function electrodes (providing concurrent stimulation and recording). Recent neurostimulators are modified to operate with these novel sensing electrodes capable of recording local field potentials (LFPs, extracellular potentials generated by the synchronized electrical activity of closely located neurons in the vicinity of an electrode [16]) together with the delivery of stimulation. This direct access to the neuronal activity domain enables the potential identification of electrophysiological biomarkers linked to pathological states of the disease (here “biomarkers” are not molecular in nature but represent instead specific signal patterns). Monitoring these biomarkers (which may be patient specific) and exploring their variability enables the adaptation of different stimulation parameters according to the needs of each patient and in response to the disease progression over time. This leads to a potential increase in the quality of life of the patient whilst lowering the cumulative effect of therapy. Unfortunately, there are still challenges that must be overcome for the successful development and implementation of electrophysiology-based closed-loop aDBS. A significant part of these challenges is related to the complex signal analysis and to the stimulation modulation.
Machine Learning (ML) methods are rapidly growing in popularity as powerful computational tools to deal with the complex data from DBS and pave the way to “smart” systems. These methods can give computers the ability to learn and improve from accumulated data (“experience”), without the need to define algorithms or specific dependencies between variables. ML offers methods to explore complex structure and patterns in the data, that are not easily identifiable with traditional methods. Given its versatility, ML can be useful at different stages of the DBS therapy process such as patient selection [17, 18], electrode placement during surgery [1921], prediction of outcome [2224], and management of stimulation [25]. The latter is linked to the identification of discriminatory electrophysiology biomarkers and associated modulation of stimulation parameters, crucial to the development of personalized and adaptive control systems for an effective symptoms relief.
The application of ML towards the development of components of an aDBS system based on electrophysiology biomarkers is the focus of this review. We discuss the current strategies and challenges towards these “smart” closed-loop aDBS technologies, and explore how ML can contribute to their development. In specific, we draw our attention to the use of ML for the identification and validation of biomarkers. PubMed was used for the literature analysis, focusing on publications with date ranging from 2015 to 2023. Further information regarding the included papers, such as the methods used and its contextualization in the DBS therapy, can be found in Tables S2–S6. In the next section, we start by providing the core concepts and terminology of ML. We then move on to exploring the search for effective biomarkers, providing fertile ground to discuss different aDBS strategies and technologies. We close the review with the discussion of the road ahead for achieving aDBS with the application of ML.

Core concepts in machine learning

Machine learning methods are a large family of data-driven algorithms capable of automatically improving their ability to make decisions/predictions through accumulated data exposure (“experience”). The application of a ML method often implies training a model with the available data, which is then tested and validated according to given performance criteria. Different metrics (defined according to the problem) are used to evaluate the performance of the model and select the most adequate for the task. There are three main types of methods used in machine learning. In supervised learning, the models are trained on annotated datasets (with labelled input–output correspondence). In unsupervised learning, methods look for hidden patterns or internal structures in the input, without pre-defined information about the output. Finally, in reinforcement learning a computational agent is trained based on policies (“rules”) that reward desired actions and punish unwanted ones. Whereas reinforcement learning methods yield a relevant potential for training neuromodulation systems, they have not yet been employed in the context of DBS for PD (but see [26]). As such, in this review we focus on unsupervised and supervised learning methods successfully used in DBS.
Unsupervised learning algorithms can help in dimensionality reduction (search for a reduced set of variables describing the data), feature extraction (identifying which variables can be helpful for discrimination and which are redundant), and clustering (grouping sets of data according to similarities or goals). Such algorithms can be used per se, to support decisions or, in combination with data representation techniques, to gain new insights (e.g. the number of classes contained in the data, how the data is distributed, and what patterns exist within the data) leading to a better selection of models in a subsequent processing stage (Fig. S1).
Supervised learning algorithms require labelled data (for every input there must be a matching annotated output) and two data subsets should be defined: a training subset, for adjusting the model’s parameters; and a testing subset, for model evaluation with data that was unseen during training (a validation subset may also be considered for adjusting the model’s hyper-parameters). This division in training data and testing data is fundamental and addresses the ability of the model to generalize, i.e. preserve adequate performance with new non-annotated data. As real-world data is noisy (from both instrumental and physiological sources), there is the risk of attaining high performance levels in the training data but low levels in the new data (a situation called overfitting). Concerning the outputs (predictions) in supervised learning, so called regression methods produce continuous values whereas classification methods are associated with categorical/discrete outputs (Fig. S1). A generic workflow to support the development of classification (and detection) methods in the context of data-driven stimulation is presented in Fig. 2.
A particular ML method called deep learning has gained considerable relevance, due to its ability to provide good estimates even when facing unstructured high dimensional data. This method is inspired by the dynamics of biological neurons and uses nodes (neuron analogues) which are combined in large architectures with several sequential layers to provide the output. The number of elements per layer, the number of layers, and the behavior of each layer (e.g., fully connected, convolutional, recurrent) are some of the parameters that can be adjusted to fit the network to the data/problem. Despite the widespread use of these techniques, the high amount of data that is required for training and the “black-box” model that is obtained in the end are strong caveats of deep neural networks. Nevertheless, visualization tools, data augmentation and transfer learning are techniques that can help solving these issues.
Both supervised and unsupervised ML methods, in a variety of different algorithms, have already been effectively used with DBS data (Table 1). In all these methods, the quality of the data deserves special attention: incorrect or poor-quality data (e.g., outliers, wrong labels, noise), if not properly cared for, will lead to under-optimized models and to unsatisfactory results. In the ML context, the discussion is often focused on the decision of the model and on the performance metrics to be considered during the development phase. In a clinical setting, the relative impact between false positive predictions and false negative predictions is an important issue on the choice of performance metrics.
Table 1
Machine learning methods in DBS
Type/purpose
Method name
Unsupervised
 
 Clustering
Gaussian mixture model
K-means
 Dimensionality reduction/feature selection
Linear discriminant analysis (LDA)
Principal component analysis (PCA): kernel PCA
t-distributed stochastic neighbour embedding (t-SNE)
Supervised
 
 Classification
AdaBoost
Decision Tree (DT): Oblique DT
Gradient Boosting Machine: XGBM, Extreme gradient boosted trees
Hidden Markov Model (HMM)
K-nearest neighbor (KNN)
Logistic Regression (LR): L1 logistic / LASSO; L2 logistic/Ridge
Naïve Bayes (NB): Conditional model and Gaussian
Neural Networks (NN): Multilayer perceptron, Shallow NN, Convolutional NN (CNN), Deep NN, LAMSTAR NN, Recurrent Networks
Random Forest (RF): unsupervised RF
Support-vector machine (SVM): SVM based on linear and Radial Basis Function (RBF) kernels
 Regression/time series
Granger causality
Linear regression
Kalman filters
Recurrent networks
Volterra kernels
The methods are organized according to the following categories: Clustering (Ct) and Dimensionality Reduction/Feature Selection (DR/FS) for unsupervised learning; Classification (Cf) and Regression/Time Series (R/TS) for supervised learning. It should be noticed that some methods can be adapted to operate with different purposes. For example, recurrent networks can be used either in a context of classification or regression
Since these approaches are "data-driven", it is essential to ensure the quality of the supporting data, used in the training phase, and to monitor the operation, in the deployment phase. Training datasets should be sufficiently diverse to guarantee that a wide range of operational situations are anticipated. The amount of data should be sufficient to ensure a reasonable fit of the model parameters, and should be balanced across classes, so that trends or decision bias do not arise. Regarding the operational phase, it is important to guarantee that safety limits are established, and that regulatory and contingency mechanisms are foreseen.

The quest for effective biomarkers

The concept of biomarkers plays a central role in the development of feedback control systems for aDBS. A biomarker refers to a characteristic in a physiological signal, such as a specific pattern or component, which correlates to a certain pathological state. Different motor or electrophysiological signals, such as electroencephalography (EEG) signals and LFPs, have shown potential for biomarker identification, and consequently closing the loop in aDBS [3, 2734]. LFPs present interesting advantages over EEG due to the source location and data fidelity, a possibility given the now available electrodes with combined stimulation and sensing properties, and appropriate long-term stability at the electrode-tissue interface [35]. Different neurological conditions have already been studied in the context of closed-loop DBS systems using LFPs as biomarkers (for an overview, see [36]).
Several aspects come into play in the identification of effective biomarkers and their selection is determined by the disease context and symptoms manifestation. Key features to consider during the selection of either single- or multi-dimensional biomarkers are the high signal-to-noise ratio of the recordings and the stability in the presence of external artifacts. These artifacts can be caused by non-pathological functions, such as intentional movement. In fact, one issue with the use of beta-band subcortical oscillations as a biomarker in PD is that they are also involved in the modulation of voluntary movement [36]. Naturally, important features also include the sensitivity and the specificity of the biomarkers, to ensure the correlation in time with the severity of the clinical symptoms [37, 38]. Independently of the source/type of the physiological signal, the search for strong correlations between signal features and pathological states is a challenging analytical task, in which ML methods can have an important contribution.

Motor biomarkers

With motor symptoms being a hallmark of PD, several studies have focused on assessing strong correlations between motor features and clinical states. Given that motility is an intrinsic feature of the human experience, it is crucial to understand what constitutes a pathological motor pattern and how it differs from normal motor activity, be it volitional or not. A common source for the study of motor data are the so called wearables: sensors that can be worn and provide detailed motor/physiological information about the patient [39]. This objective information serves as a valuable addition to the clinical data when assessing symptoms and their response to stimulation, enhancing confidence in the chosen stimulation parameters. By providing objective measurements, it also alleviates the workload on the clinicians. One of the major advantages of wearables is that they can be easily integrated in ordinary devices, such as smartwatches or rings. For instance, Powers et al. developed a motor fluctuations monitor for Parkinson’s disease algorithm, integrating long term data collection from smartwatches, capable of evaluating tremor severity and the presence of dyskinesia [40]. Additional information on movement (e.g. extracted from smartphones) can also emerge from crowd-generated data as a proxy for wearables, as shown by Zhang et al. In their work, convolutional neuronal networks (CNN) and random forest (RF) applied to smartphone accelerometer and gyroscope recordings were used to differentiate PD from control subjects [41]. Alternatively, Kleinholdermann et al. explored the use of non-invasive movement recordings to predict the best DBS settings [42]. In their work, RF models were trained to learn the relation between electrode settings, clinical ratings, and movement features (extracted from wearables). Their work shows not only that movement features were able to predict the clinical mobility (correlations up to r = 0.68 between the predicted and true values), but also that these features predicted the optimal DBS parameters (r = 0.8).
Discrimination between PD and healthy subjects is also possible using information from wearables placed on a single arm with the implementation of the K-nearest neighbors (KNN) algorithm, providing a quantitative assessment of bradykinesia, rigidity and tremor [43]. Moreover RF algorithms using 3D gait data and motor readout signals have been used to show that a standing up test can be used to distinguish PD OFF DBS patients from healthy subjects; also, foot and lower leg kinematics are better in classifying motor anomalies than other gait analysis segments [44, 45]. This contributes not only to the diagnosis of PD, but also to the monitoring of the symptom progression and response to treatment. Different approaches for monitoring symptoms, combining sensors for motion, muscle activity and force, were presented by Angeles et al. and Tahafchi et al. [46, 47]. These studies used, respectively, K-nearest neighbors and support vector machines methods to predict motor symptom severity and Freezing of Gait (FoG) events. Liu et al. took advantage of yet another RF model to classify FoG states based on the Codamotion 3-D movement capture system and synchronized intracranial EEG data, further broadening the understanding of movement dynamics and contributing to bridging the gap between peripheral and intracranial signals [48].
It is important to recall that DBS is not a substitute to pharmacologic treatment, but rather a complementary therapy. Consequently, monitoring response to medication should also be considered when designing aDBS systems. In this respect, Khodakarami et al. applied linear regression models to predict the motor symptom state based on the response to the first morning dose of levodopa [49].
The ability to detect and analyze motor patterns (directly or indirectly) may turn out to be an indispensable component of aDBS systems, capable of adjusting function over different time horizons and symptom intensities. Yet, one should not forget that the symptoms measured by wearables are but a peripheral manifestation of a neurological condition. Furthermore, the location of the wearable will affect the extracted features, and there is not a consensus on the best approach for these measurements. The information of wearables alone is, most likely, not sufficient to fulfil the needs of aDBS implementation: some of the neurological traits may not translate into noticeable motor manifestations and have non-motor implications on the patients.

Electrophysiology biomarkers

Electrophysiology signals coming from the stimulated neuronal populations can be explored either in the time domain (amplitude as a function of time) or in the frequency domain (decomposition in its frequency components; Fig. S2). The latter, often studied using spectral analysis, is generally preferred not only because it facilitates both noise and artifact identification and reduction, but also because it allows the decomposition of the signal in pre-defined frequency bands (Table S7) [37]. Variations in the power of these bands have been associated (correlated) with different PD symptoms, showing their potential to work as biomarkers for pathologic events [38]. However, accounting for the complexity of neuronal dynamics, reducing the biomarker to a frequency band’s power may leave out useful information [50]. Here, ML can help to uncover patterns that traditional signal processing tools do not.
Electrophysiology datasets (and LPFs datasets in particular) are often small, structured, and noisy. Consequently, specific techniques have been developed to circumvent these challenges. For example, these datasets are amenable to gradient-boosted tree learning, a popular ML method for prediction. Hirschmann et al. effectively used this method to demonstrate that neuronal oscillations in LFPs provide accurate predictions of motor symptom improvements following DBS [51]. In addition to signal band power, the signal coherence between subthalamic and parietal regions was shown to have relevant predictive power of DBS’ therapeutic efficacy. With their approach, it was possible to study not only the predictive capability of the LFPs, but also which features would contribute more to a potential clinical application (among the best being the sub-thalamic high beta power). Additionally, logistic regression was able to identify periods when patients have rest tremor using LFPs from the subthalamic nucleus [52]. Two studies with LFPs, using neuronal networks and hidden Markov model classifiers were also successful in decoding movement and laterality (the side in motion), as well as tremor prediction [53, 54].
Another approach to classification of human behavior with LFPs signals and considering five tasks (speech, finger movement, mouth movement, arm movement, and random segments), was possible using support vector machines (SVM) [55]. In Golshan et al., more complex methods such as CNNs were used to classify the human behavior using the time–frequency representation of STN-LFPs within the beta frequency range [56]. Interestingly, even simpler classification methods, such as the naïve Bayes classifier, can extract valuable information from LFPs. For example, LFPs dynamics on specific frequency bands (alpha and beta) allow accurate prediction of intentional limb movements, ahead of their execution [57]. Further combinations of ML techniques with Kalman filters applied to LFPs allowed the identification of biomarkers such as the power in the high beta frequency band, high gamma frequency band, and low beta frequency band for tremor detection [58].
It is also worth mentioning that potential biomarkers can be explored in a variety of neuronal activity metrics. This includes synchronization levels and long-term correlations between neuronal signals from different sources (e.g., different hemispheres or intracerebral/peripheral). For example, SVMs algorithms and Gaussian Naïve Bayes models have been applied to LFPs recordings to decode movement behavior using frequency dependent neural synchronization and inter-hemispheric connectivity features [59]. Recent literature emerged on the value of evoked resonant neural activity (ERNA) signals as potential biomarkers, however, its use is still under validation [33, 34].
Finally, it should be highlighted that the search for personalized biomarkers is gaining some traction. This is not surprising, given that the disease manifests and evolves differently in each patient. The “one size fits all” approach is being replaced by tailoring the treatment according to individual needs. Mohammed et al. implemented patient specific features extraction in combination with adaptive SVM classifiers [60]. The outcome of this method provided individualized features and a classifier for each patient. This highlights the importance to account for the heterogeneity of symptoms across patients and tones down the expectation on finding reliable population-wide neural biomarkers. On a similar note, supervised methods were used for the identification of individual neural biomarkers of hand motor performance in PD patients undergoing DBS. In this study, patient specific biomarkers improved decoding accuracy compared to group-level [61].

Other biomarkers

Besides affecting motor control, PD is also characterized by a myriad of non-motor symptoms, such as speech alteration and insomnia, highly debilitating and often hard to diagnose and manage. These symptoms could also, in principle, support relevant biomarkers. However, the identification and monitoring of non-motor symptoms poses serious challenges, due to their subjective nature. Matters become more complex if one intends to correlate these symptoms with intracranial electrophysiology signals from sensing electrodes. Regarding non-motor symptoms that can be monitored, two groups are highlighted here: speech and voice alterations, and sleep and cognition.
Speech and voice features can provide additional digital biomarkers as well as valuable information for the screening of PD patients. Given the complexity of these signals, ML methods have also been used for this type of analysis. In Braga et al., speech recordings in uncontrolled background conditions were used to identify PD patients and to estimate early signs of PD using ML regression models [62]. On another approach, by extracting paralinguistic features of voice recordings, Tracy et al. explored the possibility to predict PD severity, using both classification and regression methods. However, no clinical validation was provided in the study [63].
Within non-motor symptoms, both sleep quality and cognition worsen with progressing PD. They are particularly challenging to address, even when using ML algorithms since the collected data has a strong level of subjectivity inherent to the available assessment tools [64]. However, the study of electroencephalographic recordings shows potential to extract biomarkers for post-operative cognitive decline deterioration [65, 66]. Using LFPs recordings during wakefulness and nocturnal polysomnography sleep as inputs to SVMs and decision tree algorithms, Chen et al. showed that it is possible to classify sleep stages in PD [67].
Clinical information (admission details and UPDRS-III evaluations) has proven to be helpful in the prediction of PD progression and its response to therapy. For example, Khojandi et al. have used preoperative PD rating scores and clinical characteristics to feed a RF algorithm to predict the appropriate DBS stimulation frequency (either 60 Hz or 130–185 Hz) for a given patient [68]. In the context of PD but not directly related to DBS, Shamir et al. present a proof-of-concept of a clinical decision support system that applies ML techniques such as SVMs and RF to clinical data in order to assist in treatment management [69].
All things considered, biomarkers with high specificity and sensitivity will have a crucial role in the multiple stages of PD management: either as immediate sources of clinical information or as the means for more effective DBS protocols. Wearables, and other sources of peripheral signals related to PD symptoms, are an important strategy to provide information on the patient’s clinical state. However, the use of additional external hardware can be uncomfortable for the patient and unnecessary/redundant if neuronal (electrophysiology) biomarkers can provide equivalent information. ML techniques have been successfully applied in the study of the PD symptomatology, especially in intracranial electrophysiological signals. The ideal closed-loop control systems would probably rely solely on intracranial signals, and, in this sense, the use of LFPs related signals, captured by modern DBS systems, may be the best solution. LFP analysis should consider not only isolated values of the parameters (single or combined) but also their long-term temporal evolution and intrinsic patterns, both in physiological and pathological states. These provide a better understanding of the patient’s state, leading to the design of better control strategies. A clinically validated classification system based solely on intracranial signals has not yet been achieved, but the first steps have been taken. Although early DBS literature aimed towards the finding of generalized biomarkers for PD, recent works are increasingly adopting a personalized framework, giving primacy to individual-specific biomarkers.

Bringing adaptive DBS to the clinic

Adaptive DBS systems must simultaneously monitor (record) physiological signals and provide stimulation to the neuronal targets, while adjusting the stimulation parameters according to disease specific biomarkers reflecting the current state of the patient. It is worth mentioning that the neuronal tissue response to electrical stimulation may fluctuate over time as a result of external (environmental) or internal (disease progression) factors [70]. Efficacy of aDBS is further challenged by neuronal plasticity, which is thought to contribute to the loss of therapeutic effect over time in an open-loop framework.
Three main components are required for an aDBS system with closed-loop control using LFPs biomarkers (Fig. 4): the neurostimulator (controllable pulse generator) that provides the stimulation, a lead that delivers the stimulation and simultaneously records the LFPs, and a processing unit that reads and interprets the biomarkers present in the LFPs and decides the appropriate stimulation response. Recent technological advances in the neurostimulation devices expanded the range of tunable parameters beyond the typical pulse parameters (amplitude, frequency, and pulse-width). For example, with the availability of new leads with directional stimulation capabilities, it is also possible to shape the electrical field used in DBS [4, 7174]. All these parameters can be, a priori, usable by ML algorithms for closed-loop aDBS.

Adaptive DBS control strategies

Adaptive DBS requires Implantable Pulse Generators (IPGs) with signal sensing and recording capabilities, in addition to the stimulation delivery, in order to conduct biomarkers monitoring and generate the stimulation protocols in a real-time manner [75, 76]. Biomarkers are not required to be causally linked to the disease mechanisms, but rather to show strong correlation with symptom onset and severity, and to be sensitive or adaptive to the disease progression [7780]. Nevertheless, a causal relation would be ideal since it allows the identification of changes prior to the onset of symptoms, enabling a predictive approach rather than a responsive one. A common strategy in neuromodulation that employs preventive (and predicted) measures is the delayed feedback control method, entailing closed-loop techniques designed to desynchronize abnormal neuronal activity. Aiming at the suppression of pathological collective synchrony, effective and robust desynchronization of STN GPi model neurons has been shown by stimulation protocols based on delayed feedback control [81, 82]. The latency of the response is also an important factor to account for. Current neurotechnologies allow for virtually instantaneous and continuous adaptation of the stimulation parameter to the monitored biomarkers [83]. However, it is yet not clear how refined should the temporal resolution for aDBS be, considering the optimization of therapeutic effect and the parallel computational requirements. In Rosa et al., stimulation intensity was linearly adjusted every second, in proportion to LFP beta power. Despite only being deployed in one patient, this approach improved symptom control [84]. In Little et al., ON/OFF stimulation was adjusted continuously in a closed-loop system with improvements on motor symptoms in an eight-patient cohort [83].

Amplitude modulation

Amplitude is one of the pulse parameters that can be used to modulate the stimulation in aDBS systems [83, 85, 86] (Fig. S3). The simplest paradigm consists in the delivery of stimulation with a predefined non-null amplitude only when necessary (ON/OFF controller). Other paradigms modulate the amplitude in different ways: gradually (or, step-wise) and continuously [87]. During gradual amplitude modulation, the amplitude is maintained between two defined discrete values and varies in a step-like fashion [32, 88]. In continuous modulation, the amplitude of the stimulation varies (proportionally, in general) according to a reference signal/biomarker. The output amplitude reflects, with minimal delay, the alterations of the input signal [75, 84, 89].

Frequency and pulse-width modulation

Changes in the frequency and pulse duration of the stimulation have an impact on the neuronal response and can be used in a closed-loop context. Although the involved biological mechanisms remain unclear, high-frequency stimulation (> 100 Hz) generally produces better therapeutic results than low-frequency stimulation [90, 91]. The initial parameters for frequency and pulse-width are often in the ranges of 130–180 Hz and 60–90 µs, respectively. The fact that different symptoms respond better to specific frequency intervals is taken into account when setting parameterizations for personalized treatments [92]. Recent studies have explored the use of shorter pulse widths, which appear to offer comparable therapeutic effects with better energy management and reduced adverse effects [93]. Changes in the pulse-width have also been shown to provide clinical benefits by exciting brain sites more selectively [93, 94].

Phase modulation

A core concept supporting phase-based aDBS is the hypothesis that the precise timing of stimulation is important for its effectiveness. This is particularly relevant in the context of trying to suppress pathological collective synchronization across neuronal populations. In this approach, the stimulation signal is derived from the phase response curve of the monitored neuronal population. Under this hypothesis, adapting the stimulation at the timing that most effectively suppresses the patients tremor, appears to reduce side effects and the amount of stimulation delivered, without compromising clinical results [81, 82, 95, 96].
Concluding, one should emphasize that pioneering studies using ML to support real-time data analysis in closed-loop control strategies for aBDS in PD have already started to emerge. In Gilron et al., subcortical and cortical oscillatory signatures of motor state were used as control signals to achieve a fully embedded adaptive DBS, working outside clinical settings. Both supervised and unsupervised ML methods were implemented to assess the possibility for using (i) oscillatory phenomena in PD to decode motor fluctuations; and (ii) multiple recording sites to improve the classification of an individual’s motor state [97, 98]. In other movement disorders, such as essential tremor, ML-based stimulation protocols were already successfully applied [25]. Notwithstanding, two main concerns persist. The first has to do with the computational cost of some of these algorithms. Given the power limitations of the current processing, aDBS is advised to operate on low complexity algorithms. Secondly, the development of such system will require progressive iteration, starting from works such as the one present by Gilron et al., and learning from the protocols applied to other pathologies.

Open challenges in aDBS–future roadmap

As already explored, one of the major challenges of aDBS is the identification and validation of reliable biomarkers with high specificity and sensitivity. The recent developments in sensing technology widens the opportunity to understand how the electrophysiology at the site of stimulation evolves with the pathology. In this regard, LFPs from neurons in the stimulated regions are a prime candidate for the identification of biomarkers. For better analysis of the electrophysiological signal, novel techniques for artifact removal are important to improve the robustness of LFP-based biomarkers [99, 100]. In the established literature, attention has been devoted to the LFPs’ beta band power but, as addressed earlier, other frequency bands such as alpha and gamma also convey valuable information regarding the patient’s clinical state. Representations outside the frequency domain should also be better explored, as features in the time domain may prove useful for defining biomarkers.
Furthermore, the variability on disease and symptoms across patients motivated an increased attention on personalized therapies, where individualized biomarkers and algorithms incorporating learning capabilities are necessary. To have maximum efficacy, “smart” DBS is required to act as a personalized medicine approach. Since “smart” controllers require a training stage (either to learn from scratch or adapt), one solution is to have a period in which each patient undergoes an initial calibration for aDBS therapy, where the control strategy parameters are tuned [87]. Another factor to take into account is the importance of having a control system that is able to react to patients’ everyday actions and behaviors, automatically differentiating normal patterns from pathological activity. For both these requirements, machine learning emerges as a powerful tool to discriminate the (pathological) signatures specific for each patient, a feature hardly achievable by other methods. Still, clinical expertise remains as an irreplaceable component, especially during the initial parameterization and tuning of DBS, as symptoms exhibit rich and dynamic symptomatology in time scales that range from minutes to days/months.
Machine learning is already promoting advances in DBS [101]. However, ML methods also have limitations and constraints that should be kept in mind. Some classes of methods require large datasets and high-performance hardware for training the models, and the resulting black-box models are still met with caution. Moreover, the labelled datasets are often established in highly specific contexts, which limits the potential generalization of the extracted features. ML’s need for high quantity/high quality data can be met with sharing open and curated databases, allowing the replication and the exploration of published results in a collaborative scientific effort. In order to evaluate and compare the existing methods across studies, a consensus on the parameters by which performance is measured is needed. There is also a lack of long-term recorded data, hindering our knowledge on the long-term loss of therapeutic efficacy and rise of adverse effects. Moreover, the current computational requirements for closed-loop systems (model training, deployment, and algorithm implementation), may delay its implementation in IPGs for real-time therapy management.
The hardware requirements for closed-loop DBS running ML methods also raise attention as the novel computational features on these neurostimulators can add to the size and weight of the implant. Another challenge is the heat signature and energy consumption under their continuous activity, covering data acquisition, processing, and stimulation. Having a low power consumption balanced with rechargeable battery capacity and increased memory storage are requirements of future devices [4, 102, 103]. Currently, researchers are investigating electronic alternatives, such as memristors and other neuromorphic devices, aiming to achieve low-power, small-sized, and computationally efficient implantable devices [104]. Another comment goes to the importance of developing simulation environments and biophysical models of closed-loop stimulation to design and test aDBS systems before their implementation in vivo [105, 106].
Lastly, as ML becomes increasingly integrated into clinical practice, with aDBS being just one example, it is important to assess the impact of these methodologies on the clinical workflow. On one hand, the use of ML in therapies enhances the concept of personalized medicine but a clear understanding of these technologies is not integrated into clinical training (as it primarily falls within the realm of computational fields). By fostering a multidisciplinary environment, combining clinical and computational expertise, collaborative efforts can be made towards the effective development of aDBS. Moreover, as these technologies scale up, continuous data management and algorithm tuning become crucial, highlighting the need for a strong cooperation between clinical, engineering, and computational sciences.
To progress towards adaptive DBS, we must conceptualize it from the start as a multifactorial system. The challenges in this field are many, from hardware constraints to software needs, requirements of high amounts of data and the development of personalized models with clinical validation. Nevertheless, the technology has shown promising results and the associated improvements in the patients’ quality of life should encourage the device manufacturers to continue pushing for aDBS systems.

Conclusion

This work reviews the available research on ML implementation towards aDBS in PD. Its implementation is possible throughout all the stages of DBS therapy, and in particular towards biomarker identification/selection—that may be patient-specific—and to post-operative DBS programming, using biomarker levels and adjusting a large number of stimulation parameters. The latter may particularly benefit from ML application, due to its ability to perform complex mappings between stimulation parameters and biomarkers information. Nevertheless, there are still many open challenges that need to be addressed regarding the use of ML on aDBS. Large datasets and long-term studies, carefully planned and collected, are needed for accurate model development. The application of ML extends to the IPG hardware management whose battery duration, memory space and computational power may also benefit from its use. A key challenge is the pressing need for a better fundamental understanding of PD and of the DBS physiological mechanisms. Although ML methods provide powerful descriptive models to deal with complex data and systems, alone they are not sufficient to obtain the optimal aDBS system, presenting the need for a multidisciplinary effort, covering computational and clinical sciences. Nevertheless, these technologies have shown potential to improve patients’ quality of life. This fact alone should encourage the investment in future aDBS, not only for PD but for other neurologic disorders.

Acknowledgements

Figures 1 and 4 were created with BioRender.com.

Declarations

Conflicts of interest

On behalf of all authors, the corresponding author states that there is no conflict of interest.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

Neuer Inhalt

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat He S, Baig F, Mostofi A, Pogosyan A, Debarros J, Green AL et al (2021) Closed-loop deep brain stimulation for essential tremor based on thalamic local field potentials. Mov Disord 36(4):863–873PubMedPubMedCentralCrossRef He S, Baig F, Mostofi A, Pogosyan A, Debarros J, Green AL et al (2021) Closed-loop deep brain stimulation for essential tremor based on thalamic local field potentials. Mov Disord 36(4):863–873PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Wang CF, Yang SH, Lin SH, Chen PC, Lo YC, Pan HC et al (2017) A proof-of-principle simulation for closed-loop control based on preexisting experimental thalamic DBS-enhanced instrumental learning. Brain Stimul 10(3):672–683PubMedCrossRef Wang CF, Yang SH, Lin SH, Chen PC, Lo YC, Pan HC et al (2017) A proof-of-principle simulation for closed-loop control based on preexisting experimental thalamic DBS-enhanced instrumental learning. Brain Stimul 10(3):672–683PubMedCrossRef
4.
Zurück zum Zitat Krauss JK, Lipsman N, Aziz T, Boutet A, Brown P, Chang JW et al (2021) Technology of deep brain stimulation: current status and future directions. Nat Rev Neurol 17(2):75–97PubMedCrossRef Krauss JK, Lipsman N, Aziz T, Boutet A, Brown P, Chang JW et al (2021) Technology of deep brain stimulation: current status and future directions. Nat Rev Neurol 17(2):75–97PubMedCrossRef
6.
Zurück zum Zitat Eusebio A, Cagnan H, Brown P (2012) Does suppression of oscillatory synchronisation mediate some of the therapeutic effects of DBS in patients with Parkinson’s disease? Front Integr Neurosci 6:47PubMedPubMedCentralCrossRef Eusebio A, Cagnan H, Brown P (2012) Does suppression of oscillatory synchronisation mediate some of the therapeutic effects of DBS in patients with Parkinson’s disease? Front Integr Neurosci 6:47PubMedPubMedCentralCrossRef
7.
Zurück zum Zitat Montgomery EB, Gale JT (2008) Mechanisms of action of deep brain stimulation (DBS). Neurosci Biobehav Rev 32(3):388–407PubMedCrossRef Montgomery EB, Gale JT (2008) Mechanisms of action of deep brain stimulation (DBS). Neurosci Biobehav Rev 32(3):388–407PubMedCrossRef
8.
Zurück zum Zitat Vitek J, Zhang J, Hashimoto T, Russo G, Baker K (2012) External pallidal stimulation improves Parkinsonian motor signs and modulates neuronal activity throughout the basal ganglia thalamic network. Exp Neurol 233:581–586PubMedCrossRef Vitek J, Zhang J, Hashimoto T, Russo G, Baker K (2012) External pallidal stimulation improves Parkinsonian motor signs and modulates neuronal activity throughout the basal ganglia thalamic network. Exp Neurol 233:581–586PubMedCrossRef
9.
Zurück zum Zitat Hessa C, Vaillancourt D, Okun M (2013) The temporal pattern of stimulation may be important to the mechanism of deep brain stimulation. Bone 247:296–302 Hessa C, Vaillancourt D, Okun M (2013) The temporal pattern of stimulation may be important to the mechanism of deep brain stimulation. Bone 247:296–302
10.
Zurück zum Zitat Johnson MD, Miocinovic S, McIntyre CC, Vitek JL (2008) Mechanisms and targets of deep brain stimulation in movement disorders. Neurotherapeutics 5(2):294–308PubMedPubMedCentralCrossRef Johnson MD, Miocinovic S, McIntyre CC, Vitek JL (2008) Mechanisms and targets of deep brain stimulation in movement disorders. Neurotherapeutics 5(2):294–308PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat Obeso JA, Marin C, Rodriguez-Oroz C, Blesa J, Benitez-Temiño B, Mena-Segovia J et al (2008) The basal ganglia in Parkinson’s disease: current concepts and unexplained observations. Ann Neurol 64(SUPPL. 2):30–46 Obeso JA, Marin C, Rodriguez-Oroz C, Blesa J, Benitez-Temiño B, Mena-Segovia J et al (2008) The basal ganglia in Parkinson’s disease: current concepts and unexplained observations. Ann Neurol 64(SUPPL. 2):30–46
12.
Zurück zum Zitat Tai C-H, Boraud T, Bezard E, Bioulac B, Gross C, Benazzouz A (2003) Electrophysiological and metabolic evidence that high-frequency stimulation of the subthalamic nucleus bridles neuronal activity in the subthalamic nucleus and the substantia nigra reticulata. FASEB J 17(13):1820–1830PubMedCrossRef Tai C-H, Boraud T, Bezard E, Bioulac B, Gross C, Benazzouz A (2003) Electrophysiological and metabolic evidence that high-frequency stimulation of the subthalamic nucleus bridles neuronal activity in the subthalamic nucleus and the substantia nigra reticulata. FASEB J 17(13):1820–1830PubMedCrossRef
13.
Zurück zum Zitat Magarios-Ascone C, Pazo JH, Macadar O, Buo W (2002) High-frequency stimulation of the subthalamic nucleus silences subthalamic neurons: a possible cellular mechanism in Parkinson’s disease. Neuroscience 115(4):1109–1117CrossRef Magarios-Ascone C, Pazo JH, Macadar O, Buo W (2002) High-frequency stimulation of the subthalamic nucleus silences subthalamic neurons: a possible cellular mechanism in Parkinson’s disease. Neuroscience 115(4):1109–1117CrossRef
14.
Zurück zum Zitat Montgomery E (2004) Dynamically coupled, high-frequency reentrant, non-linear oscillators embedded in scale-free basal ganglia-thalamic-cortical networks mediating function and deep brain stimulation effects. Nonlinear Stud 11:3 Montgomery E (2004) Dynamically coupled, high-frequency reentrant, non-linear oscillators embedded in scale-free basal ganglia-thalamic-cortical networks mediating function and deep brain stimulation effects. Nonlinear Stud 11:3
15.
Zurück zum Zitat Warren G, Snyder A, Miocinovic S (2004) Deep brain stimulation creates an informational lesion of the stimulated nucleus. NeuroReport 15(7):1137–1140CrossRef Warren G, Snyder A, Miocinovic S (2004) Deep brain stimulation creates an informational lesion of the stimulated nucleus. NeuroReport 15(7):1137–1140CrossRef
16.
Zurück zum Zitat Kajikawa Y, Schroeder C (2011) How local is the local field potential? Yoshinao Neuron 72(5):847–858CrossRef Kajikawa Y, Schroeder C (2011) How local is the local field potential? Yoshinao Neuron 72(5):847–858CrossRef
17.
Zurück zum Zitat Oliveira FHM, MacHado ARP, Andrade AO (2018) On the use of t -distributed stochastic neighbor embedding for data visualization and classification of individuals with Parkinson’s disease. Comput Math Methods Med 2018:232CrossRef Oliveira FHM, MacHado ARP, Andrade AO (2018) On the use of t -distributed stochastic neighbor embedding for data visualization and classification of individuals with Parkinson’s disease. Comput Math Methods Med 2018:232CrossRef
18.
Zurück zum Zitat Castano-Candamil S, Piroth T, Reinacher P, Sajonz B, Coenen VA, Tangermann M (2019) An easy-to-use and fast assessment of patient-specific dbs-induced changes in hand motor control in Parkinson’s disease. IEEE Trans Neural Syst Rehabil Eng 27(10):2155–2163PubMedCrossRef Castano-Candamil S, Piroth T, Reinacher P, Sajonz B, Coenen VA, Tangermann M (2019) An easy-to-use and fast assessment of patient-specific dbs-induced changes in hand motor control in Parkinson’s disease. IEEE Trans Neural Syst Rehabil Eng 27(10):2155–2163PubMedCrossRef
19.
Zurück zum Zitat Kim J, Duchin Y, Shamir RR, Patriat R, Vitek J, Harel N et al (2019) Automatic localization of the subthalamic nucleus on patient-specific clinical MRI by incorporating 7 T MRI and machine learning: application in deep brain stimulation. Hum Brain Mapp 40(2):679–698PubMedCrossRef Kim J, Duchin Y, Shamir RR, Patriat R, Vitek J, Harel N et al (2019) Automatic localization of the subthalamic nucleus on patient-specific clinical MRI by incorporating 7 T MRI and machine learning: application in deep brain stimulation. Hum Brain Mapp 40(2):679–698PubMedCrossRef
20.
Zurück zum Zitat Valsky D, Blackwell K, Tamir I, Eitan R, Bergman H, Israel Z (2020) Real-time machine learning classification of pallidal borders during deep brain stimulation surgery. J Neural Eng 17(1):016021PubMedCrossRef Valsky D, Blackwell K, Tamir I, Eitan R, Bergman H, Israel Z (2020) Real-time machine learning classification of pallidal borders during deep brain stimulation surgery. J Neural Eng 17(1):016021PubMedCrossRef
21.
Zurück zum Zitat Ozturk M, Telkes I, Jimenez-Shahed J, Viswanathan A, Tarakad A, Kumar S et al (2020) Randomized, double-blind assessment of LFP versus SUA guidance in STN-DBS lead implantation: a pilot study. Front Neurosci 14:1–12CrossRef Ozturk M, Telkes I, Jimenez-Shahed J, Viswanathan A, Tarakad A, Kumar S et al (2020) Randomized, double-blind assessment of LFP versus SUA guidance in STN-DBS lead implantation: a pilot study. Front Neurosci 14:1–12CrossRef
22.
Zurück zum Zitat Farrokhi F, Buchlak QD, Sikora M, Esmaili N, Marsans M, McLeod P et al (2020) Investigating risk factors and predicting complications in deep brain stimulation surgery with machine learning algorithms. World Neurosurg 134:e325–e338PubMedCrossRef Farrokhi F, Buchlak QD, Sikora M, Esmaili N, Marsans M, McLeod P et al (2020) Investigating risk factors and predicting complications in deep brain stimulation surgery with machine learning algorithms. World Neurosurg 134:e325–e338PubMedCrossRef
23.
Zurück zum Zitat Park KH, Sun S, Lim YH, Park HR, Lee JM, Park K et al (2021) Clinical outcome prediction from analysis of microelectrode recordings using deep learning in subthalamic deep brain stimulation for Parkinson‘s disease. PLoS ONE 16:1–15 Park KH, Sun S, Lim YH, Park HR, Lee JM, Park K et al (2021) Clinical outcome prediction from analysis of microelectrode recordings using deep learning in subthalamic deep brain stimulation for Parkinson‘s disease. PLoS ONE 16:1–15
25.
Zurück zum Zitat Opri E, Cernera S, Molina R, Eisinger RS, Cagle JN, Almeida L et al (2020) Chronic embedded cortico-thalamic closed-loop deep brain stimulation for the treatment of essential tremor. Sci Trans Med 12:572CrossRef Opri E, Cernera S, Molina R, Eisinger RS, Cagle JN, Almeida L et al (2020) Chronic embedded cortico-thalamic closed-loop deep brain stimulation for the treatment of essential tremor. Sci Trans Med 12:572CrossRef
26.
Zurück zum Zitat Lu M, Wei X, Che Y, Wang J, Loparo KA (2020) Application of reinforcement learning to deep brain stimulation in a computational model of Parkinson’s disease. IEEE Trans Neural Syst Rehabil Eng 28(1):339–349PubMedCrossRef Lu M, Wei X, Che Y, Wang J, Loparo KA (2020) Application of reinforcement learning to deep brain stimulation in a computational model of Parkinson’s disease. IEEE Trans Neural Syst Rehabil Eng 28(1):339–349PubMedCrossRef
27.
Zurück zum Zitat Lettieri C, Rinaldo S, Devigili G, Pauletto G, Verriello L, Budai R et al (2012) Deep brain stimulation: subthalamic nucleus electrophysiological activity in awake and anesthetized patients. Clin Neurophysiol 123(12):2406–2413PubMedCrossRef Lettieri C, Rinaldo S, Devigili G, Pauletto G, Verriello L, Budai R et al (2012) Deep brain stimulation: subthalamic nucleus electrophysiological activity in awake and anesthetized patients. Clin Neurophysiol 123(12):2406–2413PubMedCrossRef
28.
Zurück zum Zitat Rosin B, Slovik M, Mitelman R, Rivlin-Etzion M, Haber S, Israel Z et al (2011) Closedloop deep brain stimulation is superior in ameliorating Parkinsonism. Neuron 72(2):370–384PubMedCrossRef Rosin B, Slovik M, Mitelman R, Rivlin-Etzion M, Haber S, Israel Z et al (2011) Closedloop deep brain stimulation is superior in ameliorating Parkinsonism. Neuron 72(2):370–384PubMedCrossRef
29.
Zurück zum Zitat Priori A, Foffani G, Rossi L, Marceglia S (2013) Adaptive deep brain stimulation (aDBS) controlled by local field potential oscillations. Exp Neurol 245:77–86PubMedCrossRef Priori A, Foffani G, Rossi L, Marceglia S (2013) Adaptive deep brain stimulation (aDBS) controlled by local field potential oscillations. Exp Neurol 245:77–86PubMedCrossRef
30.
Zurück zum Zitat Giannicola G, Rosa M, Servello D, Menghetti C, Carrabba G, Pacchetti C et al (2012) Subthalamic local field potentials after seven-year deep brain stimulation in Parkinson’s disease. Exp Neurol 237(2):312–317PubMedCrossRef Giannicola G, Rosa M, Servello D, Menghetti C, Carrabba G, Pacchetti C et al (2012) Subthalamic local field potentials after seven-year deep brain stimulation in Parkinson’s disease. Exp Neurol 237(2):312–317PubMedCrossRef
31.
Zurück zum Zitat Abosch A, Lanctin D, Onaran I, Eberly L, Spaniol M, Ince NF (2012) Long-term recordings of local field potentials from implanted deep brain stimulation electrodes. Neurosurgery 71(4):804–814PubMedCrossRef Abosch A, Lanctin D, Onaran I, Eberly L, Spaniol M, Ince NF (2012) Long-term recordings of local field potentials from implanted deep brain stimulation electrodes. Neurosurgery 71(4):804–814PubMedCrossRef
32.
Zurück zum Zitat Swann N, Poizner H, Houser M, Gould S, Greenhouse I, Cai W et al (2011) Deep brain stimulation of the subthalamic nucleus alters the cortical profile of response inhibition in the beta frequency band: a scalp EEG study in Parkinson’s disease. J Neurosci 31(15):5721–5729PubMedPubMedCentralCrossRef Swann N, Poizner H, Houser M, Gould S, Greenhouse I, Cai W et al (2011) Deep brain stimulation of the subthalamic nucleus alters the cortical profile of response inhibition in the beta frequency band: a scalp EEG study in Parkinson’s disease. J Neurosci 31(15):5721–5729PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Sinclair NC, McDermott HJ, Bulluss KJ, Fallon JB, Perera T, Xu SS et al (2018) Subthalamic nucleus deep brain stimulation evokes resonant neural activity. Ann Neurol 83:1027–1031PubMedPubMedCentralCrossRef Sinclair NC, McDermott HJ, Bulluss KJ, Fallon JB, Perera T, Xu SS et al (2018) Subthalamic nucleus deep brain stimulation evokes resonant neural activity. Ann Neurol 83:1027–1031PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Wiest C, He S, Duchet B, Pogosyan A, Benjaber M, Denison T et al (2023) Evoked resonant neural activity in subthalamic local field potentials reflects basal ganglia network dynamics. Neurobiol Dis 178:106019PubMedPubMedCentralCrossRef Wiest C, He S, Duchet B, Pogosyan A, Benjaber M, Denison T et al (2023) Evoked resonant neural activity in subthalamic local field potentials reflects basal ganglia network dynamics. Neurobiol Dis 178:106019PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Little S, Brown P (2012) What brain signals are suitable for feedback controzl of deep brain stimulation in Parkinson’s disease? Ann N Y Acad Sci 1265(1):9–24PubMedPubMedCentralCrossRef Little S, Brown P (2012) What brain signals are suitable for feedback controzl of deep brain stimulation in Parkinson’s disease? Ann N Y Acad Sci 1265(1):9–24PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Piña-Fuentes D, Beudel M, Little S, van Zijl J, Elting JW, Oterdoom DLM et al (2018) Toward adaptive deep brain stimulation for dystonia. Neurosurg Focus 45(2):E3PubMedCrossRef Piña-Fuentes D, Beudel M, Little S, van Zijl J, Elting JW, Oterdoom DLM et al (2018) Toward adaptive deep brain stimulation for dystonia. Neurosurg Focus 45(2):E3PubMedCrossRef
38.
Zurück zum Zitat Yin Z, Zhu G, Zhao B, Bai Y, Jiang Y, Neumann WJ et al (2021) Local field potentials in Parkinson’s disease: a frequency-based review. Neurobiol Dis 155:105372PubMedCrossRef Yin Z, Zhu G, Zhao B, Bai Y, Jiang Y, Neumann WJ et al (2021) Local field potentials in Parkinson’s disease: a frequency-based review. Neurobiol Dis 155:105372PubMedCrossRef
39.
Zurück zum Zitat LeMoyne R, Mastroianni T, Whiting D, Tomycz N (2020) Parametric evaluation of deep brain stimulation parameter configurations for Parkinson’s disease using a conformal wearable and wireless inertial sensor system and machine learning. In: IEEE, pp 3606–11 LeMoyne R, Mastroianni T, Whiting D, Tomycz N (2020) Parametric evaluation of deep brain stimulation parameter configurations for Parkinson’s disease using a conformal wearable and wireless inertial sensor system and machine learning. In: IEEE, pp 3606–11
40.
Zurück zum Zitat Powers R, Etezadi-Amoli M, Arnold E, Kianian S, Mance I, Gibiansky M et al (2021) Smartwatch inertial sensors continuously monitor real-world motor fluctuations in Parkinson’s disease. Sci Transl Med 13:7865CrossRef Powers R, Etezadi-Amoli M, Arnold E, Kianian S, Mance I, Gibiansky M et al (2021) Smartwatch inertial sensors continuously monitor real-world motor fluctuations in Parkinson’s disease. Sci Transl Med 13:7865CrossRef
41.
Zurück zum Zitat Zhang H, Deng K, Li H, Albin RL, Guan Y (2020) Deep learning identifies digital biomarkers for self-reported Parkinson’s disease. Patterns 1(3):100042PubMedPubMedCentralCrossRef Zhang H, Deng K, Li H, Albin RL, Guan Y (2020) Deep learning identifies digital biomarkers for self-reported Parkinson’s disease. Patterns 1(3):100042PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Kleinholdermann U, Bacara B, Timmermann L, Pedrosa DJ (2023) Prediction of movement ratings and deep brain stimulation parameters in idiopathic Parkinson’s disease. Neuromodulation 26:356–363PubMedCrossRef Kleinholdermann U, Bacara B, Timmermann L, Pedrosa DJ (2023) Prediction of movement ratings and deep brain stimulation parameters in idiopathic Parkinson’s disease. Neuromodulation 26:356–363PubMedCrossRef
43.
Zurück zum Zitat Huo W, Angeles P, Tai YF, Pavese N, Wilson S, Hu MT et al (2020) A Heterogeneous sensing suite for multisymptom quantification of Parkinson’s disease. IEEE Trans Neural Syst Rehabil Eng 28:1387–1406CrossRef Huo W, Angeles P, Tai YF, Pavese N, Wilson S, Hu MT et al (2020) A Heterogeneous sensing suite for multisymptom quantification of Parkinson’s disease. IEEE Trans Neural Syst Rehabil Eng 28:1387–1406CrossRef
44.
Zurück zum Zitat Kuhner A, Wiesmeier IK, Cenciarini M, Maier TL, Kammermeier S, Coenen VA et al (2020) Motion biomarkers showing maximum contrast between healthy subjects and parkinson’s disease patients treated with deep brain stimulation of the subthalamic nucleus. A Pilot Study Front Neurosci 13:1–10 Kuhner A, Wiesmeier IK, Cenciarini M, Maier TL, Kammermeier S, Coenen VA et al (2020) Motion biomarkers showing maximum contrast between healthy subjects and parkinson’s disease patients treated with deep brain stimulation of the subthalamic nucleus. A Pilot Study Front Neurosci 13:1–10
45.
Zurück zum Zitat Kuhner A, Schubert T, Cenciarini M, Wiesmeier IK, Coenen VA, Burgard W et al (2017) Correlations between motor symptoms across different motor tasks, quantified via random forest feature classification in Parkinson’s disease. Front Neurol 8:1–9CrossRef Kuhner A, Schubert T, Cenciarini M, Wiesmeier IK, Coenen VA, Burgard W et al (2017) Correlations between motor symptoms across different motor tasks, quantified via random forest feature classification in Parkinson’s disease. Front Neurol 8:1–9CrossRef
46.
Zurück zum Zitat Angeles P, Tai Y, Pavese N, Wilson S, Vaidyanathan R (2017) Automated assessment of symptom severity changes during deep brain stimulation (DBS) therapy for Parkinson’s disease. In: International Conference on Rehabilitation Robotics, pp 1512–1517 Angeles P, Tai Y, Pavese N, Wilson S, Vaidyanathan R (2017) Automated assessment of symptom severity changes during deep brain stimulation (DBS) therapy for Parkinson’s disease. In: International Conference on Rehabilitation Robotics, pp 1512–1517
47.
Zurück zum Zitat Tahafchi P, Molina R, Roper JA, Sowalsky K, Hass CJ, Gunduz A et al (2017) Freezing-of-Gait detection using temporal, spatial, and physiological features with a support-vector-machine classifier. Proc Annu Int Conf IEEE Eng Med Biol Soc EMBS 352:2867–2870 Tahafchi P, Molina R, Roper JA, Sowalsky K, Hass CJ, Gunduz A et al (2017) Freezing-of-Gait detection using temporal, spatial, and physiological features with a support-vector-machine classifier. Proc Annu Int Conf IEEE Eng Med Biol Soc EMBS 352:2867–2870
48.
Zurück zum Zitat Liu DF, Zhao BT, Zhu GY, Liu YY, Bai YT, Liu HG et al (2022) Synchronized intracranial electrical activity and gait recording in Parkinson’s disease patients with freezing of gait. Front Neurosci 16:1–11 Liu DF, Zhao BT, Zhu GY, Liu YY, Bai YT, Liu HG et al (2022) Synchronized intracranial electrical activity and gait recording in Parkinson’s disease patients with freezing of gait. Front Neurosci 16:1–11
51.
Zurück zum Zitat Hirschmann J, Steina A, Vesper J, Florin E, Schnitzler A (2022) Neuronal oscillations predict deep brain stimulation outcome in Parkinson’s disease. Brain Stimul 15(3):792–802PubMedCrossRef Hirschmann J, Steina A, Vesper J, Florin E, Schnitzler A (2022) Neuronal oscillations predict deep brain stimulation outcome in Parkinson’s disease. Brain Stimul 15(3):792–802PubMedCrossRef
52.
Zurück zum Zitat Shah SA, Tinkhauser G, Chen CC, Little S, Brown P (2018) Parkinsonian tremor detection from subthalamic nucleus local field potentials for closed-loop deep brain stimulation. In: Proceedings of Annual International Conference of the IEEE Engineering in Medicine and Biology Society, pp 2320–2324 Shah SA, Tinkhauser G, Chen CC, Little S, Brown P (2018) Parkinsonian tremor detection from subthalamic nucleus local field potentials for closed-loop deep brain stimulation. In: Proceedings of Annual International Conference of the IEEE Engineering in Medicine and Biology Society, pp 2320–2324
53.
Zurück zum Zitat Islam M, Mamun K, Deng H (2017) Decoding of human movements based on deep brain local field potentials using ensemble neural networks. Comput Intell Neurosci 176(3):139–148 Islam M, Mamun K, Deng H (2017) Decoding of human movements based on deep brain local field potentials using ensemble neural networks. Comput Intell Neurosci 176(3):139–148
54.
Zurück zum Zitat Hirschmann J, Schoffelen JM, Schnitzler A, van Gerven MAJ (2017) Parkinsonian rest tremor can be detected accurately based on neuronal oscillations recorded from the subthalamic nucleus. Clin Neurophysiol 128(10):2029–2036PubMedCrossRef Hirschmann J, Schoffelen JM, Schnitzler A, van Gerven MAJ (2017) Parkinsonian rest tremor can be detected accurately based on neuronal oscillations recorded from the subthalamic nucleus. Clin Neurophysiol 128(10):2029–2036PubMedCrossRef
55.
Zurück zum Zitat Golshan H, Hebb A, Hanrahan S, Nedrud J, Mahoor MH, Golshan HM et al (2018) A hierarchical structure for human behavior classification using STN local field potentials. J Neurosci Methods 293:254–263PubMedCrossRef Golshan H, Hebb A, Hanrahan S, Nedrud J, Mahoor MH, Golshan HM et al (2018) A hierarchical structure for human behavior classification using STN local field potentials. J Neurosci Methods 293:254–263PubMedCrossRef
56.
Zurück zum Zitat Golshan HM, Hebb AO, Mahoor MH (2020) LFP-Net: a deep learning framework to recognize human behavioral activities using brain STN-LFP signals. J Neurosci Methods 335:108621PubMedCrossRef Golshan HM, Hebb AO, Mahoor MH (2020) LFP-Net: a deep learning framework to recognize human behavioral activities using brain STN-LFP signals. J Neurosci Methods 335:108621PubMedCrossRef
57.
Zurück zum Zitat Khawaldeh S, Tinkhauser G, Shah SA, Peterman K, Debove I, Khoa Nguyen TA et al (2020) Subthalamic nucleus activity dynamics and limb movement prediction in Parkinson’s disease. Brain 143(2):582–586PubMedPubMedCentralCrossRef Khawaldeh S, Tinkhauser G, Shah SA, Peterman K, Debove I, Khoa Nguyen TA et al (2020) Subthalamic nucleus activity dynamics and limb movement prediction in Parkinson’s disease. Brain 143(2):582–586PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Yao L, Brown P, Shoaran M (2020) Improved detection of Parkinsonian resting tremor with feature engineering and Kalman filtering. Clin Neurophysiol 131(1):274–284PubMedCrossRef Yao L, Brown P, Shoaran M (2020) Improved detection of Parkinsonian resting tremor with feature engineering and Kalman filtering. Clin Neurophysiol 131(1):274–284PubMedCrossRef
59.
Zurück zum Zitat Mamun KA, Mace M, Lutman ME, Stein J, Liu X, Aziz T et al (2015) Movement decoding using neural synchronization and inter-hemispheric connectivity from deep brain local field potentials. J Neural Eng 12(5):056011PubMedCrossRef Mamun KA, Mace M, Lutman ME, Stein J, Liu X, Aziz T et al (2015) Movement decoding using neural synchronization and inter-hemispheric connectivity from deep brain local field potentials. J Neural Eng 12(5):056011PubMedCrossRef
60.
Zurück zum Zitat Mohammed A, Zamani M, Bayford R, Demosthenous A (2015) Patient specific Parkinson’s disease detection for adaptive deep brain stimulation. In: Proceedings of Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBS), pp 1528–1531 Mohammed A, Zamani M, Bayford R, Demosthenous A (2015) Patient specific Parkinson’s disease detection for adaptive deep brain stimulation. In: Proceedings of Annual International Conference of the IEEE Engineering in Medicine and Biology Society (EMBS), pp 1528–1531
61.
Zurück zum Zitat Castaño-Candamil S, Piroth T, Reinacher P, Sajonz B, Coenen VA, Tangermann M (2020) Identifying controllable cortical neural markers with machine learning for adaptive deep brain stimulation in Parkinson’s disease. NeuroImage Clin 28:102376PubMedPubMedCentralCrossRef Castaño-Candamil S, Piroth T, Reinacher P, Sajonz B, Coenen VA, Tangermann M (2020) Identifying controllable cortical neural markers with machine learning for adaptive deep brain stimulation in Parkinson’s disease. NeuroImage Clin 28:102376PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Braga D, Madureira AM, Coelho L, Ajith R (2018) Automatic detection of Parkinson’s disease based on acoustic analysis of speech. Eng Appl Artif Intell 2019(77):148–158 Braga D, Madureira AM, Coelho L, Ajith R (2018) Automatic detection of Parkinson’s disease based on acoustic analysis of speech. Eng Appl Artif Intell 2019(77):148–158
63.
Zurück zum Zitat Tracy JM, Özkanca Y, Atkins DC, Hosseini GR (2019) Investigating voice as a biomarker: Deep phenotyping methods for early detection of Parkinson’s disease. J Biomed Inform 2020(104):103362 Tracy JM, Özkanca Y, Atkins DC, Hosseini GR (2019) Investigating voice as a biomarker: Deep phenotyping methods for early detection of Parkinson’s disease. J Biomed Inform 2020(104):103362
65.
Zurück zum Zitat Geraedts VJ, Koch M, Kuiper R, Kefalas M, Bäck THW, van Hilten JJ et al (2021) Preoperative electroencephalography-based machine learning predicts cognitive deterioration after subthalamic deep brain stimulation. Mov Disord 36(10):2324–2334PubMedPubMedCentralCrossRef Geraedts VJ, Koch M, Kuiper R, Kefalas M, Bäck THW, van Hilten JJ et al (2021) Preoperative electroencephalography-based machine learning predicts cognitive deterioration after subthalamic deep brain stimulation. Mov Disord 36(10):2324–2334PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Geraedts VJ, Koch M, Contarino MF, Middelkoop HAM, Wang H, van Hilten JJ et al (2021) Machine learning for automated EEG-based biomarkers of cognitive impairment during deep brain stimulation screening in patients with Parkinson’s Disease. Clin Neurophysiol 132(5):1041–1048PubMedCrossRef Geraedts VJ, Koch M, Contarino MF, Middelkoop HAM, Wang H, van Hilten JJ et al (2021) Machine learning for automated EEG-based biomarkers of cognitive impairment during deep brain stimulation screening in patients with Parkinson’s Disease. Clin Neurophysiol 132(5):1041–1048PubMedCrossRef
67.
Zurück zum Zitat Chen Y, Gong C, Hao H, Guo Y, Xu S, Zhang Y et al (2019) Automatic sleep stage classification based on subthalamic local field potentials. IEEE Trans Neural Syst Rehabil Eng 27(2):118–128PubMedPubMedCentralCrossRef Chen Y, Gong C, Hao H, Guo Y, Xu S, Zhang Y et al (2019) Automatic sleep stage classification based on subthalamic local field potentials. IEEE Trans Neural Syst Rehabil Eng 27(2):118–128PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Khojandi A, Shylo O, Mannini L, Kopell BH, Ramdhani RA (2017) Stratifying Parkinson’s patients with STN-DBS into high-frequency or 60 Hz-frequency modulation using a computational model. Neuromodulation 20(5):450–455PubMedCrossRef Khojandi A, Shylo O, Mannini L, Kopell BH, Ramdhani RA (2017) Stratifying Parkinson’s patients with STN-DBS into high-frequency or 60 Hz-frequency modulation using a computational model. Neuromodulation 20(5):450–455PubMedCrossRef
69.
Zurück zum Zitat Shamir R, Dolber T, Noecker A, Walter B, McIntyre C (2015) Machine learning approach to optimizing combined stimulation and medication therapies for Parkinson’s disease. Brain Stimul 8(6):1025–1032PubMedPubMedCentralCrossRef Shamir R, Dolber T, Noecker A, Walter B, McIntyre C (2015) Machine learning approach to optimizing combined stimulation and medication therapies for Parkinson’s disease. Brain Stimul 8(6):1025–1032PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Hunka K, Suchowersky O, Wood S, Derwent L, Kiss Z (2005) Nursing time to program and assess deep brain stimulators in movement disorder patients. J Neurosci Nurs 37:204–210PubMedCrossRef Hunka K, Suchowersky O, Wood S, Derwent L, Kiss Z (2005) Nursing time to program and assess deep brain stimulators in movement disorder patients. J Neurosci Nurs 37:204–210PubMedCrossRef
71.
Zurück zum Zitat Bour LJ, Lourens MAJ, Verhagen R, De Bie RMA, Van Den Munckhof P, Schuurman PR et al (2015) Directional recording of subthalamic spectral power densities in Parkinson’s disease and the effect of steering deep brain stimulation. Brain Stimul 8(4):730–741PubMedCrossRef Bour LJ, Lourens MAJ, Verhagen R, De Bie RMA, Van Den Munckhof P, Schuurman PR et al (2015) Directional recording of subthalamic spectral power densities in Parkinson’s disease and the effect of steering deep brain stimulation. Brain Stimul 8(4):730–741PubMedCrossRef
72.
Zurück zum Zitat Schnitzler A, Mir P, Brodsky MA, Verhagen L, Groppa S, Alvarez R et al (2021) Directional deep brain stimulation for Parkinson’s disease: results of an international crossover study with randomized, double-blind primary endpoint. Neuromodulation 25(6):817–828CrossRef Schnitzler A, Mir P, Brodsky MA, Verhagen L, Groppa S, Alvarez R et al (2021) Directional deep brain stimulation for Parkinson’s disease: results of an international crossover study with randomized, double-blind primary endpoint. Neuromodulation 25(6):817–828CrossRef
73.
Zurück zum Zitat Sabourin S, Khazen O, DiMarzio M, Staudt MD, Williams L, Gillogly M et al (2020) Effect of directional deep brain stimulation on sensory thresholds in Parkinson’s disease. Front Hum Neurosci 14:1–8CrossRef Sabourin S, Khazen O, DiMarzio M, Staudt MD, Williams L, Gillogly M et al (2020) Effect of directional deep brain stimulation on sensory thresholds in Parkinson’s disease. Front Hum Neurosci 14:1–8CrossRef
74.
Zurück zum Zitat Rammo RA, Ozinga SJ, White A, Nagel SJ, Machado AG, Pallavaram S et al (2021) Directional stimulation in Parkinson’s disease and essential tremor: the Cleveland clinic experience. Neuromodulation 25(6):829–835CrossRef Rammo RA, Ozinga SJ, White A, Nagel SJ, Machado AG, Pallavaram S et al (2021) Directional stimulation in Parkinson’s disease and essential tremor: the Cleveland clinic experience. Neuromodulation 25(6):829–835CrossRef
75.
Zurück zum Zitat Arlotti M, Marceglia S, Foffani G, Volkmann J, Lozano AM, Moro E et al (2018) Eight-hours adaptive deep brain stimulation in patients with Parkinson disease. Neurology 90(11):e971–e976PubMedPubMedCentralCrossRef Arlotti M, Marceglia S, Foffani G, Volkmann J, Lozano AM, Moro E et al (2018) Eight-hours adaptive deep brain stimulation in patients with Parkinson disease. Neurology 90(11):e971–e976PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Sarica C, Iorio-Morin C, Aguirre-Padilla D, Najjar A, Paff M, Fomenko A et al (2021) Implantable pulse generators for deep brain stimulation: challenges, complications, and strategies for practicality and longevity. Front Hum Neurosci 15:489CrossRef Sarica C, Iorio-Morin C, Aguirre-Padilla D, Najjar A, Paff M, Fomenko A et al (2021) Implantable pulse generators for deep brain stimulation: challenges, complications, and strategies for practicality and longevity. Front Hum Neurosci 15:489CrossRef
77.
Zurück zum Zitat Kühn AA, Kupsch A, Schneider GH, Brown P (2006) Reduction in subthalamic 8–35 Hz oscillatory activity correlates with clinical improvement in Parkinson’s disease. Eur J Neurosci 23(7):1956–1960PubMedCrossRef Kühn AA, Kupsch A, Schneider GH, Brown P (2006) Reduction in subthalamic 8–35 Hz oscillatory activity correlates with clinical improvement in Parkinson’s disease. Eur J Neurosci 23(7):1956–1960PubMedCrossRef
78.
Zurück zum Zitat Neumann W, Staub-Bartelt F, Horn A, Schanda J, Schneider G, Brown P et al (2017) Long term correlation of subthalamic beta band activity with motor impairment in patients with Parkinson’s disease. Clin Neurophysiol 128(11):2286–2291PubMedPubMedCentralCrossRef Neumann W, Staub-Bartelt F, Horn A, Schanda J, Schneider G, Brown P et al (2017) Long term correlation of subthalamic beta band activity with motor impairment in patients with Parkinson’s disease. Clin Neurophysiol 128(11):2286–2291PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat Wang DDD, Hemptinne C, Miocinovic S, Qasim S, Miller A, Ostrem J et al (2016) Subthalamic local field potentials in Parkinson’s disease and isolated dystonia: an evaluation of potential biomarkers. Neurobiol Dis 89:213–222PubMedPubMedCentralCrossRef Wang DDD, Hemptinne C, Miocinovic S, Qasim S, Miller A, Ostrem J et al (2016) Subthalamic local field potentials in Parkinson’s disease and isolated dystonia: an evaluation of potential biomarkers. Neurobiol Dis 89:213–222PubMedPubMedCentralCrossRef
80.
Zurück zum Zitat De Hemptinne C, Ryapolova-Webb ES, Air EL, Garcia PA, Miller KJ, Ojemann JG et al (2013) Exaggerated phase-amplitude coupling in the primary motor cortex in Parkinson disease. Proc Natl Acad Sci USA 110(12):4780–4785PubMedPubMedCentralCrossRef De Hemptinne C, Ryapolova-Webb ES, Air EL, Garcia PA, Miller KJ, Ojemann JG et al (2013) Exaggerated phase-amplitude coupling in the primary motor cortex in Parkinson disease. Proc Natl Acad Sci USA 110(12):4780–4785PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Popovych OV, Lysyansky B, Rosenblum M, Pikovsky A, Tass PA (2017) Pulsatile desynchronizing delayed feedback for closed-loop deep brain stimulation. PLoS ONE 12(3):1–29CrossRef Popovych OV, Lysyansky B, Rosenblum M, Pikovsky A, Tass PA (2017) Pulsatile desynchronizing delayed feedback for closed-loop deep brain stimulation. PLoS ONE 12(3):1–29CrossRef
82.
Zurück zum Zitat Popovych OV, Lysyansky B, Tass PA (2017) Closed-loop deep brain stimulation by pulsatile delayed feedback with increased gap between pulse phases. Sci Rep 7(1):1–14CrossRef Popovych OV, Lysyansky B, Tass PA (2017) Closed-loop deep brain stimulation by pulsatile delayed feedback with increased gap between pulse phases. Sci Rep 7(1):1–14CrossRef
83.
Zurück zum Zitat Little S, Pogosyan A, Neal S, Zavala B, Zrinzo L, Hariz M et al (2013) Adaptive deep brain stimulation in advanced Parkinson disease. Ann Neurol 74(3):449–457PubMedPubMedCentralCrossRef Little S, Pogosyan A, Neal S, Zavala B, Zrinzo L, Hariz M et al (2013) Adaptive deep brain stimulation in advanced Parkinson disease. Ann Neurol 74(3):449–457PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Rosa M, Arlotti M, Ardolino G, Cogiamanian F, Marceglia S, Di Fonzo A et al (2015) Adaptive deep brain stimulation in a freely moving parkinsonian patient. Mov Disord 30(7):1003–1005PubMedPubMedCentralCrossRef Rosa M, Arlotti M, Ardolino G, Cogiamanian F, Marceglia S, Di Fonzo A et al (2015) Adaptive deep brain stimulation in a freely moving parkinsonian patient. Mov Disord 30(7):1003–1005PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Khobragade N, Graupe D, Tuninetti D (2015) Towards fully automated closed-loop deep brain stimulation in parkinson’s disease patients: a LAMSTAR-based tremor predictor. In: Proceedings of IEEE Engineering in Medicine and Biology Society, pp 2616–2619 Khobragade N, Graupe D, Tuninetti D (2015) Towards fully automated closed-loop deep brain stimulation in parkinson’s disease patients: a LAMSTAR-based tremor predictor. In: Proceedings of IEEE Engineering in Medicine and Biology Society, pp 2616–2619
86.
Zurück zum Zitat Basu I, Graupe D, Tuninetti D, Shukla P, Slavin KV et al (2013) Pathological tremor prediction using surface electromyogram and acceleration: potential use in ‘ON–OFF’ demand driven deep brain stimulator design. J Neural Eng 10(3):036019PubMedPubMedCentralCrossRef Basu I, Graupe D, Tuninetti D, Shukla P, Slavin KV et al (2013) Pathological tremor prediction using surface electromyogram and acceleration: potential use in ‘ON–OFF’ demand driven deep brain stimulator design. J Neural Eng 10(3):036019PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Vissani M, Isaias IU, Mazzoni A (2020) Deep brain stimulation: a review of the open neural engineering challenges. J Neural Eng 17(5):051002PubMedCrossRef Vissani M, Isaias IU, Mazzoni A (2020) Deep brain stimulation: a review of the open neural engineering challenges. J Neural Eng 17(5):051002PubMedCrossRef
88.
Zurück zum Zitat Malekmohammadi M, Herron J, Velisar A, Blumenfeld Z, Trager MH, Chizeck HJ et al (2016) Kinematic adaptive deep brain stimulation for resting tremor in Parkinson’s disease. Mov Disord 31(3):426–428PubMedCrossRef Malekmohammadi M, Herron J, Velisar A, Blumenfeld Z, Trager MH, Chizeck HJ et al (2016) Kinematic adaptive deep brain stimulation for resting tremor in Parkinson’s disease. Mov Disord 31(3):426–428PubMedCrossRef
89.
Zurück zum Zitat Rosa M, Arlotti M, Marceglia S, Cogiamanian F, Ardolino G, Fonzo Di A et al (2017) Adaptive deep brain stimulation controls levodopa-induced side effects in Parkinsonian patients. Mov Disord 32(4):628–629PubMedPubMedCentralCrossRef Rosa M, Arlotti M, Marceglia S, Cogiamanian F, Ardolino G, Fonzo Di A et al (2017) Adaptive deep brain stimulation controls levodopa-induced side effects in Parkinsonian patients. Mov Disord 32(4):628–629PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Baizabal-Carvallo JF, Alonso-Juarez M (2016) Low-frequency deep brain stimulation for movement disorders. Park Relat Disord 31:14–22CrossRef Baizabal-Carvallo JF, Alonso-Juarez M (2016) Low-frequency deep brain stimulation for movement disorders. Park Relat Disord 31:14–22CrossRef
92.
Zurück zum Zitat Koeglsperger T, Palleis C, Hell F, Mehrkens J, Bötzel K (2019) Deep brain stimulation programming for movement disorders: current concepts and evidence-based strategies. Front Neurol 10:410PubMedPubMedCentralCrossRef Koeglsperger T, Palleis C, Hell F, Mehrkens J, Bötzel K (2019) Deep brain stimulation programming for movement disorders: current concepts and evidence-based strategies. Front Neurol 10:410PubMedPubMedCentralCrossRef
93.
Zurück zum Zitat Steigerwald F, Timmermann L, Kühn A, Schnitzler A, Reich MM, Kirsch AD et al (2018) Pulse duration settings in subthalamic stimulation for Parkinson’s disease. Mov Disord 33(1):165–169PubMedCrossRef Steigerwald F, Timmermann L, Kühn A, Schnitzler A, Reich MM, Kirsch AD et al (2018) Pulse duration settings in subthalamic stimulation for Parkinson’s disease. Mov Disord 33(1):165–169PubMedCrossRef
94.
Zurück zum Zitat Akbar U, Raike RS, Hack N, Hess CW, Skinner J, Martinez-Ramirez D et al (2016) Randomized, blinded pilot testing of nonconventional stimulation patterns and shapes in Parkinson’s disease and essential tremor: evidence for further evaluating narrow and biphasic pulses. Neuromodulation Technol Neural Interface 19(4):343–356CrossRef Akbar U, Raike RS, Hack N, Hess CW, Skinner J, Martinez-Ramirez D et al (2016) Randomized, blinded pilot testing of nonconventional stimulation patterns and shapes in Parkinson’s disease and essential tremor: evidence for further evaluating narrow and biphasic pulses. Neuromodulation Technol Neural Interface 19(4):343–356CrossRef
95.
Zurück zum Zitat Popovych OV, Tass PA (2010) Synchronization control of interacting oscillatory ensembles by mixed nonlinear delayed feedback. Phys Rev E 82(2):1–7CrossRef Popovych OV, Tass PA (2010) Synchronization control of interacting oscillatory ensembles by mixed nonlinear delayed feedback. Phys Rev E 82(2):1–7CrossRef
96.
Zurück zum Zitat Popovych OV, Hauptmann C, Tass PA (2005) Effective desynchronization by nonlinear delayed feedback. Phys Rev Lett 94(16):2–5CrossRef Popovych OV, Hauptmann C, Tass PA (2005) Effective desynchronization by nonlinear delayed feedback. Phys Rev Lett 94(16):2–5CrossRef
97.
Zurück zum Zitat Gilron R, Little S, Perrone R, Wilt R, de Hemptinne C, Yaroshinsky MS et al (2021) Long-term wireless streaming of neural recordings for circuit discovery and adaptive stimulation in individuals with Parkinson’s disease. Nat Biotechnol 39:1078–1085PubMedPubMedCentralCrossRef Gilron R, Little S, Perrone R, Wilt R, de Hemptinne C, Yaroshinsky MS et al (2021) Long-term wireless streaming of neural recordings for circuit discovery and adaptive stimulation in individuals with Parkinson’s disease. Nat Biotechnol 39:1078–1085PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Sarikhani P, Ferleger B, Mitchell K, Ostrem J, Herron J, Mahmoudi B et al (2022) Automated deep brain stimulation programming with safety constraints for tremor suppression in patients with Parkinson’s disease and essential tremor. J Neural Eng 19(4):046042CrossRef Sarikhani P, Ferleger B, Mitchell K, Ostrem J, Herron J, Mahmoudi B et al (2022) Automated deep brain stimulation programming with safety constraints for tremor suppression in patients with Parkinson’s disease and essential tremor. J Neural Eng 19(4):046042CrossRef
99.
Zurück zum Zitat Rossi L, Foffani G, Marceglia S, Bracchi F, Barbieri S, Priori A (2007) An electronic device for artefact suppression in human local field potential recordings during deep brain stimulation. J Neural Eng 4(2):96–106PubMedCrossRef Rossi L, Foffani G, Marceglia S, Bracchi F, Barbieri S, Priori A (2007) An electronic device for artefact suppression in human local field potential recordings during deep brain stimulation. J Neural Eng 4(2):96–106PubMedCrossRef
100.
Zurück zum Zitat Stanslaski S, Afshar P, Cong P, Giftakis J, Stypulkowski P, Carlson D et al (2012) Design and validation of a fully implantable, chronic, closed-loop neuromodulation device with concurrent sensing and stimulation. IEEE Trans Neural Syst Rehabil Eng 20(4):410–421PubMedCrossRef Stanslaski S, Afshar P, Cong P, Giftakis J, Stypulkowski P, Carlson D et al (2012) Design and validation of a fully implantable, chronic, closed-loop neuromodulation device with concurrent sensing and stimulation. IEEE Trans Neural Syst Rehabil Eng 20(4):410–421PubMedCrossRef
101.
Zurück zum Zitat Peralta M, Jannin P, Baxter J (2021) Machine learning in deep brain stimulation: a systematic review. Artif Intell Med 122:102198PubMedCrossRef Peralta M, Jannin P, Baxter J (2021) Machine learning in deep brain stimulation: a systematic review. Artif Intell Med 122:102198PubMedCrossRef
102.
Zurück zum Zitat Verma N, Shoeb A, Bohorquez J, Dawson J, Guttag J, Chandrakasan AP (2010) A micro-power EEG acquisition SoC with integrated feature extraction processor for a chronic seizure detection system. IEEE J Solid-State Circuits 45(4):804–816CrossRef Verma N, Shoeb A, Bohorquez J, Dawson J, Guttag J, Chandrakasan AP (2010) A micro-power EEG acquisition SoC with integrated feature extraction processor for a chronic seizure detection system. IEEE J Solid-State Circuits 45(4):804–816CrossRef
103.
Zurück zum Zitat Dams J, Siebert U, Bornschein B, Volkmann J, Deuschl G, Oertel WH et al (2013) Cost-effectiveness of deep brain stimulation in patients with Parkinson’s disease. Mov Disord 28(6):763–771PubMedCrossRef Dams J, Siebert U, Bornschein B, Volkmann J, Deuschl G, Oertel WH et al (2013) Cost-effectiveness of deep brain stimulation in patients with Parkinson’s disease. Mov Disord 28(6):763–771PubMedCrossRef
104.
Zurück zum Zitat Dias C, Castro D, Aroso M, Ventura J, Aguiar P (2022) A memristor-based neuromodulation device for real-time monitoring and adaptive control of in vitro neuronal populations. ACS Appl Electron Mater 4(5):2380–2387PubMedPubMedCentralCrossRef Dias C, Castro D, Aroso M, Ventura J, Aguiar P (2022) A memristor-based neuromodulation device for real-time monitoring and adaptive control of in vitro neuronal populations. ACS Appl Electron Mater 4(5):2380–2387PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Jovanov I, Nauman M, Kumaravelu K, Lesi V, Zutshi A, Grill WM et al (2018) Learning-based control design for deep brain stimulation. In: Proceedings of 9th ACM/IEEE International Conference on Cyber-Physical Systems (ICCPS), pp 349–3450 Jovanov I, Nauman M, Kumaravelu K, Lesi V, Zutshi A, Grill WM et al (2018) Learning-based control design for deep brain stimulation. In: Proceedings of 9th ACM/IEEE International Conference on Cyber-Physical Systems (ICCPS), pp 349–3450
Metadaten
Titel
Machine learning for adaptive deep brain stimulation in Parkinson’s disease: closing the loop
verfasst von
Andreia M. Oliveira
Luis Coelho
Eduardo Carvalho
Manuel J. Ferreira-Pinto
Rui Vaz
Paulo Aguiar
Publikationsdatum
02.08.2023
Verlag
Springer Berlin Heidelberg
Erschienen in
Journal of Neurology / Ausgabe 11/2023
Print ISSN: 0340-5354
Elektronische ISSN: 1432-1459
DOI
https://doi.org/10.1007/s00415-023-11873-1

Weitere Artikel der Ausgabe 11/2023

Journal of Neurology 11/2023 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Hyponatriämie Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.

Schutz der Synapsen bei Alzheimer

29.05.2024 Morbus Alzheimer Nachrichten

Mit einem Neurotrophin-Rezeptor-Modulator lässt sich möglicherweise eine bestehende Alzheimerdemenz etwas abschwächen: Erste Phase-2-Daten deuten auf einen verbesserten Synapsenschutz.

Sozialer Aufstieg verringert Demenzgefahr

24.05.2024 Demenz Nachrichten

Ein hohes soziales Niveau ist mit die beste Versicherung gegen eine Demenz. Noch geringer ist das Demenzrisiko für Menschen, die sozial aufsteigen: Sie gewinnen fast zwei demenzfreie Lebensjahre. Umgekehrt steigt die Demenzgefahr beim sozialen Abstieg.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.