Skip to main content

Anatomo-physiological basis and applied techniques of electrical neuromodulation in chronic pain

Abstract

Chronic pain, a complex and debilitating condition, poses a significant challenge to both patients and healthcare providers worldwide. Conventional pharmacological interventions often prove inadequate in delivering satisfactory relief while carrying the risks of addiction and adverse reactions. In recent years, electric neuromodulation emerged as a promising alternative in chronic pain management. This method entails the precise administration of electrical stimulation to specific nerves or regions within the central nervous system to regulate pain signals. Through mechanisms that include the alteration of neural activity and the release of endogenous pain-relieving substances, electric neuromodulation can effectively alleviate pain and improve patients' quality of life. Several modalities of electric neuromodulation, with a different grade of invasiveness, provide tailored strategies to tackle various forms and origins of chronic pain. Through an exploration of the anatomical and physiological pathways of chronic pain, encompassing neurotransmitter involvement, this narrative review offers insights into electrical therapies’ mechanisms of action, clinical utility, and future perspectives in chronic pain management.

Introduction

The International Association for the Study of Pain (IASP) defines pain as "an unpleasant sensory and emotional experience associated with, or resembling that associated with, actual or potential tissue damage" [1]. In 2013, the IASP established a task force to develop and update a classification of pain disorders. This effort led to the inclusion of a chronic pain classification in the 2019 edition of the International Classification of Diseases (ICD-11), formally adopted by the World Health Organization [2, 3]. Chronic pain is defined by a duration of at least three months or beyond the usual healing period. It can be further categorized into nociceptive, neuropathic, and nociplastic pain types [4]. Chronic pain presents a significant public health challenge, affecting millions of patients and incurring substantial medical expenses and lost productivity [5, 6]. According to the Global Burden of Disease Study, tension-type headache, migraine, low back pain, neck pain, diabetic neuropathy are among the most common and prevalent chronic pain syndromes in the population, increasing their years lived with disability [6].

In a certain percentage of patients, conventional treatments, including surgery, pharmacological therapies, combined with psychological, physical and occupational therapies, fail [7,8,9]. In these patients, neuromodulation may have a role in treatment. Neuromodulation refers to a broad category of techniques or therapies that modulate the activity of the nervous system to achieve therapeutic effects through chemical (i.e. pharmacological) interventions or electrical stimulation (i.e. neuromodulation). Neurostimulation, which involve modifying or stimulating nerve activity through targeted electrical at specific neurological sites, is increasingly utilized in patients with chronic pain of varied origins [10]. These modalities represent the evolving landscape for chronic pain management, each offering unique advantages and considerations [11].

This review aims to provide a comprehensive understanding of the neuroanatomy and neurophysiology of chronic pain, with an emphasis on the role of electrical neuromodulation in its treatment.

Neuroanatomy and neurophysiology of chronic pain

Pain pathways are intricate and dynamic systems comprising sensory, cognitive, and behavioral components. These mechanisms have evolved to recognize, integrate, and coordinate protective responses to noxious stimuli. They encompass both primitive spinal reactions and the nuanced emotional experiences consciously identified as pain in humans [12].

In addition to the peripheral pain pathways, which include receptors and neural fibers, the central nervous system (CNS) pain pathways involve a network of structures. These structures comprise the spinal cord, thalamus, amygdala, hypothalamus, periaqueductal grey (PAG) matter, basal ganglia, insular and cingulate cortices, as well as the sensory and motor cortices [13]. Figure 1 illustrates a schematic representation of pain pathways.

Fig. 1
figure 1

Anatomical pathways of chronic pain and neurotransmitters. The figure depicts the ascending and descending anatomical pathways of pain modulation. In the inner panel, neurotransmitters (depicted as squares) and receptors (depicted as V shapes) are illustrated. These include GABA (red), glycine (black), opiates (brown), norepinephrine (gray), glutamate (green), substance P (light blue), and serotonin (violet), each with their respective inhibitory (-) or excitatory ( +) activity indicated

Pain signaling from periphery to ascending pathways

The intricate process of a painful stimulus begins in the periphery with specialized nociceptors activated by stimuli like pressure, extreme temperatures, or tissue damage. Nociceptors transmit the pain stimulus to primary afferent fibers, responsible for carrying sensory information from the periphery to the CNS. Classified based on diameter and conduction velocity, two basic types of nerve fibers exist: myelinated Type A and unmyelinated Type C. Pain transmission occurs through two types: rapid (associated with Aδ-fibers) and slow (related to polymodal receptors and C fibers), each transmitting pain at different speeds. The cell bodies of sensory neurons are situated in the dorsal root ganglia (DRG); these neurons are capable of encoding and transmitting information derived from external stimuli. As these primary afferent fibers approach the spinal cord, they enter through the dorsal roots (i.e., dorsal root entry zone) [12, 13]. Inside the spinal cord, unmyelinated and small myelinated axons project laterally to enter the Lissauer tract, a marginal zone or layer of the dorsal horn. Here, they synapse with neurons in the dorsal horn. Aδ fibers ascend three to four segments in the Lissauer tract before terminating in the Rexed’s lamina I, II outer, or V. In contrast, C fibers typically ascend one segment before ending in lamina II.

One of the principal conduits in this relay of pain signals is the spinothalamic tract. This major ascending pathway serves as a dedicated highway for transmitting pain signals to the brain. The second-order neurons of the spinothalamic tract, crucial intermediaries in this relay, undergo a remarkable anatomical phenomenon known as decussation. These neurons cross over to the contralateral side of the spinal cord before ascending towards the thalamus, a central hub for sensory processing [14]. The thalamus acts as a sensory relay station, receives the pain signals, and orchestrates their distribution to various cortical areas. From the thalamus, projections fan out to regions of the brain responsible for the conscious perception of pain. These cortical areas, intricately connected and functioning in concert, give rise to the multifaceted experience of pain that encompasses its sensory, emotional, and cognitive dimensions [14].

The spinoreticular tract, diverging from traditional sensory pathways, projects to the reticular formation in the brainstem, influencing emotional and autonomic aspects of pain perception. Additionally, the spinocerebellar tract, known for proprioceptive transmission, also contributes to nociception and motor responses to pain, further intertwining sensory perception and motor output [13].

Pain modulation from descending pathways

As pain signals ascend from the periphery to the brain through the intricate pathways, a parallel network of descending modulatory pathways operates in concert, finely tuning the transmission and perception of pain, maintaining a delicate balance between the intensity of noxious stimuli and the perceptual experience of pain.

One of the most prominent players in the descending modulatory orchestra is the descending inhibitory pathway. This pathway exerts its influence through a series of neural structures, i.e. the PAG, situated around the cerebral aqueduct in the midbrain, and the rostroventral medulla (RVM), a region placed in the brainstem. These regions control the release of endogenous opioids and other neurotransmitters that act as pain inhibitors [15, 16]. In particular, the PAG integrates information from various brain regions; its activation largely reduces perception of pain [17].

Descending from the PAG, the inhibitory signals make their way to the RVM. The RVM acts as a relay station, forwarding the inhibitory commands to the spinal cord. Within the spinal cord, these commands target nociceptive neurons, the primary relay stations for pain signals. The release of endogenous opioids, such as enkephalins and endorphins, is a hallmark of this inhibitory process [18]. These opioids bind to receptors on nociceptive neurons, effectively dampening their activity and reducing the transmission of pain signals. Beyond endogenous opioids, neurotransmitters like serotonin and norepinephrine also play crucial roles in the descending inhibitory pathway. The release of these neurotransmitters contributes to the overall suppression of nociceptive signals. Serotonin is known for its involvement in pain modulation and mood regulation, highlighting the intricate interplay between sensory and emotional components in pain perception[18].

The descending inhibitory pathway not only reduces the transmission of pain signals but also contributes to the phenomenon of "pain gating". The Gate Control Theory, proposed by Melzack and Wall, suggests that non-nociceptive inputs can modulate pain perception [19].

The dorsal column of the spinal cord comprises various neural fibers, with certain types such as Aδ and C fibers being triggered by painful stimuli, while the larger Aβ fibers serve to suppress the transmission of these signals. According to the "Gate Control Theory," the interplay between the activity of Aδ and C fibers versus that of Aβ fibers dictates the perception of painful stimuli [7].

RVM is also known for the presence of "on-cells," which release neurotransmitters such as Substance P, glutamate, and norepinephrine. These neurotransmitters have the effect of amplifying the activity of nociceptive neurons in the spinal cord rather than inhibiting them [18].

Neurotransmitters of pain

Multiple neurotransmitters participate in both transmitting and modulating pain signals, exerting effects that can either enhance or suppress the perception of pain [20].

Substance P, a neuropeptide functioning as an excitatory neurotransmitter, is primarily synthesized and secreted by nociceptive neurons, transmitting and modulating pain signals throughout the CNS [21]. In the spinal cord, Substance P is released both at synaptic junctions and non-synaptic sites within the dorsal horn. It binds to tachykinins (NK1, NK2 and NK3) receptors located on lamina 1 neurons and the dendrites of lamina 5 neurons [22]. This interaction facilitates the transmission of pain signals from peripheral nerves to the CNS and assists in relaying these signals to higher brain regions responsible for processing pain perception. Additionally, Substance P can induce the release of other neurotransmitters, such as glutamate, thereby intensifying the pain signals [23]. Moreover, Substance P contributes to the emotional and affective dimensions of pain perception, influencing brain areas associated with mood regulation, stress responses, and emotional processing [24].

Glutamate is another excitatory neurotransmitter in the CNS involved in the conduction of pain signals by activating amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid (AMPA) and N-Methyl-D-Aspartate (NMDA) receptors on postsynaptic neurons in the dorsal horn of the spinal cord [25]. This activation induces the depolarization of postsynaptic neurons, generating action potentials and transmitting pain signals along ascending pathways to the brain centers [26]. NMDA receptors are crucial for amplifying and modulating pain signals, especially in chronic pain conditions. Unlike AMPA receptors, NMDA receptors necessitate both glutamate binding and postsynaptic depolarization for full activation of their ion channels. This property allows NMDA receptors to integrate synaptic activity and contribute to the phenomenon of central sensitization, wherein pain signals are amplified and prolonged in chronic pain conditions [27].

Calcitonin Gene-Related Peptide (CGRP) is a neuropeptide synthetized and released from sensory nerves in response to noxious stimuli, inflammation, and tissue injury. CGRP is a potent vasodilator, and it contributes to inflammation and hypersensitivity [28]. In addition, it enhances the excitability of sensory neurons and promotes the release of other neurotransmitters, such as substance P [29]. CGRP has received significant attention in the context of migraine since elevated levels have been found in the blood and cerebrospinal fluid during attacks. Therefore, CGRP has become a target for novel treatments for these patients [30].

Norepinephrine is released from descending pathways (particularly from the locus coeruleus), having both inhibitory and excitatory effects on pain transmission. Its net effect depends on the receptors it acts upon, with α2-adrenergic receptors generally inhibiting pain signals [31, 32]. Direct stimulation of the PAG or RVM elevates norepinephrine levels in the cerebrospinal fluid, then modulating pain transmission in the spinal cord by inhibition of the release of glutamate and substance P [33].

Serotonin, also known as 5-hydroxytryptamine (5-HT), is another neurotransmitter involved in pain modulation. It exhibits both excitatory and inhibitory effects on pain transmission, which are contingent upon the receptor subtype activated and the neural pathways engaged [34]. 5-HT exerts its effects through multiple receptor subtypes (including 5-HT1, 5-HT2, 5-HT3, and so on), differently distributed throughout the nervous system and with distinct functional properties. Of note, 5-HT is characterized by an excitatory or inhibitory effect on pain transmission according to the activated subtype of receptors [35]. For example, 5-HT2 and 5-HT3 receptors enhance pain transmission by increasing neuronal excitability and amplifying the signaling of pain pathways. Conversely, 5-HT1 receptors suppress the release of excitatory neurotransmitters, dampen neuronal activity, and reduce the transmission of pain signals, resulting in pain relief [34]. It's worth noting that in the typical condition, 5-HT concentrations are relatively low, with a certain level of facilitation mediated by 5-HT3 and 5-HT2 receptors. Subsequent elevations in spinal 5-HT then result in the inhibitory actions of the 5-HT7 receptor [35].

5-HT exerts regulatory effects on pain pathways across various levels of the nervous system. Apart from its role in the spinal cord, it also impacts pain processing in several brain regions associated with pain and anxiety, including the thalamus, frontal cortex, amygdala, and brainstem [36].

Gamma-Aminobutyric Acid (GABA) is the principal inhibitory neurotransmitter in the CNS. GABA produces its inhibitory effects by inducing hyperpolarization of postsynaptic neurons, thereby increasing the negativity of the neuron's membrane potential. This hyperpolarization reduces the likelihood of the neuron generating an action potential, leading to decreased excitability and limiting the propagation of signals along the neural pathway [37]. Within pain modulation, GABAergic activity mitigates pain signals, regulating both the intensity and duration of pain perception [24]. In the spinal cord, GABA is released onto postsynaptic neurons subsequent to receiving inputs from primary nociceptive neurons, exerting inhibitory control [38]. Conversely, GABA modulates various regions in the brain such as the thalamus, where it inhibits transmission to the cortex, as well as the somatosensory cortex, insula, and anterior cingulate cortex. Furthermore, GABA influences descending pain pathways including the PAG and RVM [39]. Additionally, GABAergic inhibition extends to the emotional and affective dimensions of pain processing, impacting brain regions like the amygdala and prefrontal cortex, thus modulating emotions such as fear, anxiety, and depression [24].

Endorphins and enkephalins, endogenous opioids synthesized in diverse CNS locales such as the hypothalamus, pituitary gland, and spinal cord, serve as neurotransmitters and neuromodulators, predominantly triggered by stress, pain, and physical exertion [40]. A key function of these compounds is pain signal inhibition, facilitated by their binding to mu (μ), delta (δ), and kappa (κ) opioid receptors on neurons in the spinal cord and brain, initiating intracellular cascades that lead to neuronal suppression [41]. Within the spinal cord, endorphins and enkephalins hinder the release of substance P and glutamate while diminishing the excitability of pain-transmitting neurons, thereby modulating the transmission of pain signals from the periphery to the brain. Moreover, these opioids exert their analgesic effects across various brain regions, including the PAG, RVM, thalamus, and limbic system [42].

Peripheral and central sensitization

Peripheral sensitization is a condition defined by the IASP as a state of “Increased responsiveness and diminished threshold of nociceptive neurons in the periphery to the stimulation of their receptive fields” [43, 44]. This phenomenon occurs because of chemical mediators released by nociceptors and various non-neuronal cells, such as mast cells, basophils, platelets, macrophages, neutrophils, endothelial cells, keratinocytes, and fibroblasts, at the site of tissue injury or inflammation. A plethora of signaling molecules is involved in this process, including protons, ATP, prostaglandins (PGE2), thromboxanes, leukotrienes, endocannabinoids, growth factors (neurotrophins, granulocyte- or granulocyte-macrophage colony-stimulating factors), cytokines (IL6, IL1β, TNFα), chemokines, neuropeptides (CGRP, substance P, bradykinin, histamine), lipids, and various proteases [45,46,47,48,49].

Conversely, central sensitization is a complex phenomenon defined by an amplification of neural signaling within the CNS that elicits pain hypersensitivity [50]. In particular, it is marked by lasting changes in the excitability of second-order neurons within the spinal cord, induced by increased afferent activity. This results in significant alterations to the somatosensory system itself [51]. The profound significance of this concept was emphasized by the description of the long-term potentiation in the hippocampus, revealing that synchronous high-frequency input enhances synaptic efficacy [52]. Central sensitization has been postulated to contribute in several chronic pain syndromes, such as rheumatoid arthritis, osteoarthritis, temporomandibular disorders, fibromyalgia, musculoskeletal disorders, headache, neuropathic pain, complex regional pain syndrome [50, 53].

Neurostimulation techniques

The fundamental principle of neurostimulation in chronic pain revolves around precisely delivering electrical impulses to modulate nervous system activity, thereby modifying pain perception, and providing relief. This approach aims to interrupt or dampen pain signals along neural pathways, by either stimulating specific nerves to block pain transmission or influencing the brain's processing of pain signals [54]. More precisely, in the complexity of anatomical and physiological mechanisms of chronic pain, neurostimulation, in its various forms, interferes with the hyperexcitability of circuits involved in pain processing (such as the spinal cord stimulation and peripheral nerve stimulation) [55, 56], enhances endogenous pain inhibition (such as the motor cortex stimulation and the transcranial magnetic stimulation) [57,58,59], or modulates the affective component of pain [54, 60, 61]. Neurostimulation is an alternative or adjunctive approach to manage chronic pain, particularly when medical or surgical treatments have been ineffective or associated with undesirable side effects [54].

Neurostimulation therapies can be classified by invasiveness. Invasive methods involve surgical implantation of electrodes or devices, including motor cortex stimulation, deep brain stimulation, vagus nerve stimulation, spinal cord stimulation, and peripheral nerve stimulation (Figure 2). Minimally invasive modalities, like pulsed radiofrequency therapy and percutaneous electrical nerve stimulation are less intrusive but involve some intervention (Figure 3). Lastly, noninvasive techniques, such as transcranial direct current stimulation, transcranial magnetic stimulation, transcutaneous electrical nerve stimulation, transcranial focused ultrasound, do not require any surgical intervention or implantation and are typically administered externally to the body (Figure 3) [54].

Fig. 2
figure 2

Invasive neurostimulation techniques. The invasive techniques for neurostimulation of chronic pain are depicted alongside their current indications with clear ( +) or unclear ( ±) evidence. The specific characteristics of each modality are explained in the text

Fig. 3
figure 3

Mini-invasive and non-invasive neurostimulation techniques. The mini-invasive (i.e. pulsed radiofrequency therapy and percutaneous electrical nerve stimulation) and non-invasive (i.e. transcranial direct current stimulation, transcranial magnetic stimulation, transcutaneous electrical nerve stimulation and transcranial focused ultrasound) techniques for neurostimulation of chronic pain are schematized in the representation alongside their current indications with clear ( +) or unclear ( ±) evidence. The specific characteristics of each modality are explained in the text

Motor cortex stimulation

Motor Cortex Stimulation (MCS) consists in the implantation of an array of electrodes directly onto the motor cortex of the brain contralateral to the painful side [62, 63]. Particularly, after craniotomy, the electrode array is positioned in the precentral gyrus or central sulcus corresponding to the painful area either in the epidural or subdural spaces [64, 65]. Although there is not an evidence-based difference, opting for subdural lead placement may be more reasonable due to the reduced distance between the leads and the brain cortex compared to epidural placement [66].

MCS is theorized to alleviate pain by influencing both the emotional dimension of pain and inhibiting pain signals at different neural levels. Specifically, MCS relieves pain by activating the perigenual cingulate and orbitofrontal cortex, thus modulating the emotional component of pain. Simultaneously, MCS inhibits pain impulses at the spinal cord level by activating the PAG [67].

The research of MCS as a potential treatment for various neuropathic chronic pain populations has yielded inconsistent results. This variability is primarily attributable to small sample sizes and study designs of lower quality, suggesting that MCS is efficient only in specific subgroups of patients. Noteworthy, MCS has demonstrated effectiveness in addressing chronic pain associated with phantom limb pain, trigeminal neuropathic pain, complex regional pain syndrome, central post-stroke pain, brachial plexus avulsion, postsurgical pain, and pain resulting from spinal cord injury [66, 68,69,70,71,72,73,74].

Deep brain stimulation

Deep Brain Stimulation (DBS) consists in the surgical implantation of thin electrodes within specific area of the brain to be stimulated. In case of chronic pain, electrodes are generally positioned in the PAG, in the periventricular gray matter, the ventralposterior medial or the ventral posterior lateral nuclei of the thalamus (contralateral to the side of pain), the anterior cingulate cortex or the posterior insula [75, 76]. The positioning generally required a frame-based or frame-less stereotactic technique, targeting the area with a magnetic resonance imaging on the day before surgery and, in some centers, with intraoperative microelectrode recordings. Electrodes are then connected to an impulse generator that neuromodulate the target area [77].

When PAG or thalamus are stimulated, DBS seems to promote the secretions of endogenous opioids (i.e. beta-endorphin and methionine enkephalin) [78, 79]. Beside these reports, the other underlying mechanisms are still obscure.

Clinical studies have demonstrated a significant improvement in patients with central post-stroke pain [80, 81], in chronic pain associated with Parkinson’s disease [82, 83] and in those patients with spinal cord injury [84]. The most favorable outcomes in DBS, characterized by effects surpassing 50%, were observed when stimulating the somatosensory thalamus in individuals experiencing peripheral neuropathic pain [85, 86]. Nevertheless, the current trend indicates a decline in the number of patients undergoing implantation, primarily attributed to the emergence of new and less invasive treatment modalities. In addition, although considered safe, DBS is affected by adverse event in 8–9% of patients, including lead fractures, wound infections, intra-operative seizure and postoperative burr hole site erosion [87]. It is noteworthy that individuals exhibiting severe, debilitating neuropathic pain accompanied by verifiable pathology, resistant to conventional treatments, appear to be the most suitable candidates for this approach.

Vagus nerve stimulation

Vagus Nerve Stimulation (VNS) is a therapeutic method that employs electrical impulses to activate the vagus nerve, a component of the autonomic nervous system. Although widely recognized for its efficacy in treating epilepsy [88] and depression [89], VNS has also been investigated in chronic pain [90]. VNS involves the surgical placement of a stimulator device, delivering regular electrical impulses, connected to electrodes stimulating the vagus nerve. More recently, transcutaneous VNS and minimally invasive form of percutaneous VNS have been also developed [90].

The vagus nerve’s afferent pathway initiates from vagal afferents that supply the cervical, thoracic, and abdominal organs. Primarily traversing the nucleus of the solitary tract, it ultimately reaches higher brain regions, including the thalamus, hypothalamus, parabrachial nucleus, PAG, amygdala, and locus coeruleus [91,92,93,94]. The stimulation of vagus nerve modulates the descending serotoninergic and noradrenergic neurons, modulating the central sensitization and reducing the pain transmission [90]. VNS also induces anti-inflammatory effects through diverse pathways, such as the cholinergic anti-inflammatory pathway, the hypothalamic–pituitary–adrenal axis pathway, and the production of specialized pro-resolving mediators. Specifically, VNS initiates the release of acetylcholine from Vagus Nerve efferents in the coeliac ganglia and norepinephrine from the splenic nerve, subsequently promoting further release of acetylcholine [95]. Following its release, acetylcholine binds to the α7 nicotinic Ach receptors present on macrophages, leading to a decrease in cytokine production [95, 96]. Additionally, VNS activates the hypothalamic–pituitary–adrenal axis, triggering the release of adrenocorticotropic hormone. This hormone, in turn, acts on the adrenal glands, promoting the synthesis of cortisol [97]. Furthermore, VNS has the potential to induce the production of specialized pro-resolving mediators [98], which act on receptors expressed on immune cells, gliacytes, and neurons, thereby exerting a modulatory effect on inflammation and neuroinflammation [99,100,101].

Therapeutic effects on chronic pain have been casually discovered during the application of VNS for epilepsy. Some epileptic patients reported a reduction in headache frequency and intensity that after VNS implantation [102,103,104]. Following these first reports, several studies demonstrated the efficiency of VNS in preventing and reducing the intensity of headache, either with invasive implantation [105] and during transcutaneous stimulation [106,107,108,109]. Given the efficacy in modulation of central sensitization [90] and in depressive disease [89], VNS was reported to successfully reduce symptoms in patients with fibromyalgia [110], although these results were not further confirmed by other investigations [111]. VNS has been also investigated in other scenarios of chronic neuropathic pain (i.e. trigeminal allodynia and diabetic peripheral neuropathic pain) in animal models [112,113,114,115]. However, data in patients still lack and clinical studies are needed to validate the positive findings reported by animal studies.

Spinal cord stimulation

Spinal Cord Stimulation (SCS) is a neuromostimulation technique designed to address and alleviate neuropathic chronic pain. This method involves an implanted generator that produces pulsed electrical signals, which are then delivered to a specific area of the spinal cord through electrodes implanted within the epidural space [7]. Advancements in SCS techniques and equipment are ongoing, with dedicated literature available elsewhere [7]. SCS operates through neurophysiological and neurochemical mechanisms rooted in the "gate control theory” for pain transmission [7, 19].

SCS disrupts the processing of nociceptive signals through the lateral spinothalamic tract, influencing supraspinal brain centers like the ventral posterior nucleus of the thalamus, somatosensory cortex, cingulate cortex, and insula [116, 117]. Orthodromically, SCS depolarizes Aβ fibers cranially, controlling supraspinal centers such as the cuneate and gracile nuclei [118, 119]. After supraspinal integration, descending feedback loops from the locus coeruleus [120], nucleus raphe magnus [121], and rostral ventromedial medulla [122] modulate and control the spinal nociceptive signal at the "spinal gate" via serotonergic and noradrenergic projections to the dorsal horn [118, 119].

In terms of neurochemical mechanisms, SCS modulates gamma-aminobutyric acid (GABA) [123], serotonin [124], acetylcholine and norepinephrine [125, 126]. Studies in animal models show increased intraspinal release of GABA [127, 128] and attenuation of glutamate and aspartate responses [129]. Notably, GABA type "b" receptors play a crucial role, suggesting the potential for intrathecal baclofen administration to enhance SCS analgesia [130]. SCS also elevates serotonin and substance P release [124], enhances dynorphin and enkephalin expression [131], and decreases neuronal excitability and spinal pain transmission by activating 5-HT2A, 5-HT3, and 5-HT4 receptors [132]. Additionally, SCS promotes analgesia by modulating cholinergic and adrenergic neurotransmission, releasing acetylcholine and noradrenaline in the dorsal horn of the spinal cord [125, 126].

SCS is considered for those patients experiencing chronic pain, particularly when other conventional treatments (including pharmacological therapies, surgical treatments or physiotherapy) have proven ineffective or are associated with intolerable side effects. To define the indication of SCS implantation, the selection of the patient is fundamental for its success [133]. Firstly, SCS should be contemplated within two years of symptom onset, following the ineffectiveness of all standard therapies [134, 135]. It should be mentioned that the time between initial pain diagnosis and SCS implantation is still debated. In a study by Kumar et al., a time to SCS treatment < 2 years was characterized by a higher long term success rate (around 85%), declining precipitously at the lengthening of the diagnosis to implantation interval [136]. Another study by the same group reported that the greatest improvement in pain relief was obtained in case of SCS implantation within one year from the onset of symptoms [137]. However, several studies have shown a symptom-to-implantation waiting time from 5 to 6 years [138, 139]. Secondly, individuals with underlying psychiatric conditions, complete cognitive impairment, psychological comorbidities, or substance abuse are ineligible for SCS implantation [140]. However, if there is partial cognitive impairment, SCS may be considered, with a preference for non-rechargeable over rechargeable implantable pulse generators [134]. Thirdly, SCS is a viable option for neuropathic pain conditions (e.g., failed back surgery syndrome, arachnoiditis, complex regional pain syndrome, causalgia, peripheral neuropathy, chronic radiculopathy), while its efficacy is limited for nociceptive symptoms or central neuropathic pain [134]. SCS is strongly recommended in specific scenarios, including failed back surgery syndrome without neurologic progression [141], axial low back pain [142] and complex regional pain syndrome [143]. Furthermore, SCS is recommended for chronic refractory angina unresponsive to maximal medical therapy, bypass surgery, and percutaneous angioplasty of the legs [144], as well as for peripheral artery disease or non-reconstructable critical leg ischemia [145]. In particular, SCS is employed in patients experiencing chronic leg ischemia that is not suitable for open surgical or endovascular intervention [146]. This condition is typically characterized by either rest pain alone (Fontaine stage III) or the presence of rest pain accompanied by arterial ulcers less than 3 cm in diameter (Fontaine stage IV) [147].

Dorsal root ganglion stimulation

Dorsal root ganglion (DRG) stimulation resembles SCS, but with devices specifically design to target the first-order sensory neurons located in the DRG. The DRG's advantageous position within the epidural space, immersed in cerebral spinal fluid, makes it an ideal target for stimulation, allowing precise targeting of chronic pain. DRG stimulation applies an electrical field directly over the nuclei of primary afferent neurons, enabling modulation prior to signal propagation within the spinal cord. This modulation potentially includes inhibition at synapses with second-order neurons in the dorsal horn [148].

Recent pre-clinical and clinical research is shedding light on the mechanisms and therapeutic effects of DRG stimulation, highlighting differences compared to SCS. Unlike SCS, DRG stimulation does not elevate the inhibitory GABA neurotransmitter in either the dorsal horn or the DRG itself [149, 150]. The most accepted mechanism of DRG stimulation involves impeding the transmission of afferent pain signals and ectopic action potential transmission, supported by basic science research [151,152,153]. However, its effects on multi-dermatomal conditions with a single lead placement may depend on orthodromic propagation, a mechanism often discussed in SCS but less so in DRG stimulation [154, 155]. DRG stimulation is believed to activate Aδ-, Aβ-, and C-fiber low threshold mechanoreceptor fibers, utilizing the endogenous opioid system to modulate touch and pain processes at clinically utilized frequencies [156,157,158]. Furthermore, DRG stimulation has effects on the sympathetic nervous system, including antidromic effects in the treatment of peripheral vascular disease, reduction in blood pressure, and attenuation of neuroinflammation [159,160,161,162].

In 2019, the Neuromodulation Appropriateness Consensus Committee highlighted the significant efficacy of DRG stimulation in treating both complex regional pain syndrome types I and II, as well as focal neuropathic pain with identifiable pathology [163]. In fact, a large prospective randomized controlled trial, comparing SCS to DRG stimulation in patients affected by complex regional pain syndrome types I and II, demonstrated that 81.2% of DRG patients achieved ≥ 50% pain relief compared to 55.7% in the SCS arm [164].

Today the DRG stimulation is mainly accepted for the treatment of complex regional pain syndrome types I and II, although it has been attempted in several other forms of neuropathic chronic pain (including painful diabetic neuropathy, mononeuropathy, polyneuropathy) with for these latter a low-quality study and limited evidence [148, 165].

Peripheral nerve stimulation

Peripheral Nerve Stimulation (PNS) consists in the delivery of electrical impulses through electrodes surgically implanted near peripheral nerves, to modulate the transmission of pain signals along these nerves [166]. As per the SCS, PNS engages neurophysiological and neurochemical mechanisms based on the "gate control theory" of pain transmission, as above discussed [7, 19].

PNS can be implanted in various anatomical regions, including the upper extremities (brachial plexus, suprascapular nerve, axillary nerve, radial nerve, median nerve, or ulnar nerve), lower extremities (sciatic nerve, obturator nerve, femoral nerve, lateral femoral cutaneous nerve, genicular nerve, saphenous nerve, common peroneal nerve, tibial nerve, sural nerve, and superficial peroneal nerve), as well as other nerves in the head (occipital nerve, trigeminal nerve, and supraorbital nerve), trunk, abdomen, back, or pelvis (medial branch nerve, ilioinguinal nerve, iliohypogastric nerve, genitofemoral nerve, cluneal nerve, and pudendal nerve) [166].

The available evidence on PNS in the head and neck region has predominantly centered in case reports or small case series around the stimulation of occipital nerves for the treatment of headache disorders with a positive therapeutic effect [167,168,169,170]. On the opposite, the evidence for facial pain is still weak, lacking high quality studies [171,172,173,174]. Indeed, the current guidelines do not recommend PNS for chronic facial pain, whereas they state that PNS can be considered in case of chronic migraine headache after failure of conventional treatments [175].

According to the guidelines [175], PNS can also be considered as a treatment option for mononeuropathies affecting the upper extremity, failed back surgery syndrome, and chronic low back pain (supported by substantial evidence). Additionally, it may be explored in cases of radiculopathy and post-herpetic neuralgia, although the evidence for these conditions is limited. Furthermore, PNS shows promise in managing neuropathic pain in the lower extremities, including post-amputation pain, as well as in individuals with Complex Regional Pain Syndrome Type I/II or peripheral causalgia (while acknowledging that SCS remains the preferred neurostimulation option) [175].

Pulsed radiofrequency therapy

Pulsed Radiofrequency Therapy (PRF) is a medical intervention employed in the management of chronic pain conditions. In contrast to continuous radiofrequency, which subjects target nerves or tissues to sustained high temperatures (70–90 °C) through electrical stimulation [176], PRF utilizes electrical pulses delivered to specific nerves via an electrode generating localized thermal effects without causing substantial tissue damage [177]. Specifically, PRF employs a radiofrequency current characterized by alternately repeated electrical stimulation with a short duration (around 20 ms) followed by a resting phase (around 480 ms). This alternating pattern induces a temperature on the target tissue that remains below 42 °C [178].

PRF induces a targeted and prolonged reduction in spinal sensitization mediated by C-fibers. As a result, it effectively inhibits the transmission of pain signals from peripheral nerves to the central nervous system [179, 180]. PRF is reversible and temporary, it provides relief for a variable duration, and the procedure may be repeated if necessary [178].

PRF is commonly applied to peripheral nerves, DRG, or other nerve structures contributing to chronic pain. Therefore, PRF is often used to modulate neural activity and disrupt pain signals without causing tissue damage, whereas continuous radiofrequency creates thermal lesions that disrupt nerve function and alleviate pain [176,177,178]. Based on these different effects, PRF is commonly employed for conditions where nerve-related pain is the primary concern, such as neuropathic pain syndromes or neuralgias. On the opposite, continuous radiofrequency is frequently used for conditions characterized by localized pain arising from specific anatomical structures, such as facet joints or sacroiliac joints [176,177,178].

The most important efficacy of PRF has been shown in the treatment of postherpetic neuralgia [181] targeting the areas near the DRG via the angulus costae [182] or paravertebral puncture [183] or targeting the intercostal nerves [184, 185].

In recent findings, the literature has indicated limited and weak evidence for the use of PRF on the lumbar DRG for radicular pain [186]; in facets’ pain, PRF has been also considered to be inferior to continuous radiofrequency and it should be used only in selected cases [187]. Similar conclusions have been also drawn for cervical radicular pain and lumbar face pain [187]. On the opposite, intradiscal PRF seems to be a promising technique in case of discogenic pain, radiculitis, or spinal stenosis, although further studies are still required to confirm this indication [187].

Percutaneous electrical nerve stimulation

Percutaneous Electrical Nerve Stimulation (PENS) is a procedure using thin needles inserted through the skin to deliver electrical impulses to specific nerves or tissues [188]. The objective is to alleviate persistent and chronic pain by stimulation of peripheral sensory nerves through the insertion of needles in the outer layers of the skin, characterized by high resistance [188].

Today, the evidence in favor of PENS is quite weak. PENS (alone or in adjunct to other treatments) was shown to not provide a sufficient advantage in the control of musculoskeletal pain [189]. In one study including 17 patients with neuropathic pain due to spinal cord injury, PENS reduced the symptoms in 63% of cases [190]. However, no other studies have been conducted so far in this population, and the indication still lacks clear evidence. Another trial conducted in a population of patients with chronic low back pain secondary to degenerative disk disease, PENS was more effective in reducing pain and post-treatment function, as compared to Transcutaneous Electrical Nerve Stimulation (TENS) or other exercise therapies [191]. Finally, in a randomized double-blind crossover trial on 31 patients with neuropathic chronic pain, preliminary findings indicated that PENS may offer effective short-term pain relief [192].

Unfortunately, the underlying mechanisms of action of PENS are still few investigated, although they are thought to rely in the Gate Control Theory [7]. Despite its potential benefits in pain management, the precise indications for PENS, as well as the most effective treatment parameters, remain topics of discussion and investigation within the medical community. Additionally, comparative studies evaluating PENS against other therapeutic modalities are limited, leaving uncertainties about its relative efficacy and place in treatment algorithms [189, 193, 194]. To address these gaps in knowledge, future research efforts should prioritize well-designed studies that investigate the clinical utility of PENS across various pain conditions. These studies should aim to delineate the specific patient populations most likely to benefit from PENS, identify optimal stimulation parameters, and compare its efficacy and safety to other established treatments [189, 193, 194]. Furthermore, incorporating mechanistic studies to elucidate the underlying pathways through which PENS exerts its therapeutic effects can provide valuable insights into its mode of action and potential advantages over alternative interventions.

Transcranial direct current stimulation

Transcranial Direct Current Stimulation (tDCS) involves the non-invasive delivery of low-intensity electrical currents to specific brain regions for a duration of 20–30 min. These currents induce polarity-dependent shifts in resting membrane potential, thereby modulating neuronal activity at the site of stimulation and its associated structures [195, 196]. Typically, two electrodes, one anode positioned over the primary motor cortex and the other a cathode over the contralateral supraorbital region, are placed on the scalp. This setting generates a current flow through the prefrontal cortex, the cingulate cortex, the insula, and deeper structures such as thalamic and brainstem nuclei. The current flow modulates the rest membrane potential of axons, promoting the alleviation of pain [197].

To date, the efficacy of tDCS is limited to treatment of chronic neuropathic pain secondary to spinal cord injury [195] or for peripheral neuropathic pain [198]. Another recent meta-analysis has demonstrated that tDCS does not reduce the pain and related disability in non-specific chronic low-back pain syndrome, limiting the support for its use in this field [199]. Furthermore, a recent systematic review has assessed the efficacy of tDCS in migraine, reporting a positive effect on treatment success, although the included studies were quite heterogeneous and the sample size was small; therefore, these limitations precluded any definitive conclusion on the real efficacy of tDCS in this field of application [200].

Transcranial magnetic stimulation

Repetitive Transcranial Magnetic Stimulation (rTMS) is a non-invasive neurostimulation technique, applying a magnetic field externally to the scalp to induce electrical currents in targeted regions of the brain beneath the coil [201]. TMS operates on Faraday's principle of electromagnetic induction, where a rapid electric current passing through a coil for a brief duration, typically 1 ms, generates a potent electromagnetic field [202]. In TMS, the magnetic field is painlessly administered to the scalp, and cortical axons serve as the electrical conductors that receive the induced electric current [202].

The analgesic effect is obtained by stimulating the primary motor cortex (precentral gyrus), with a stimulation frequency ranging between 5 to 20 Hz, contralaterally to the side of pain localization [203, 204]. Therefore, rTMS has the capacity to regulate cortical hyperexcitability and pain pathways, including descending inhibitory pathways [205, 206]. These pathways exert an influence on supraspinal pain tracts and regions of the brain associated with social-affective functions, such as the right temporal lobe [207, 208].

rTMS has been applied in different populations of patients with chronic pain. In patients with migraine, rTMS decreases the intensity of the attack soon after the treatment application, particularly in those patients with cephalgia after traumatic brain injury [209]. High frequency rTMS on motor cortex has also proved to provide a positive temporary effect on pain in patients with neuropathic chronic pain correlated to phantom limb [210] and to manage pain alongside cognition and sleep disturbances in patients of fibromyalgia [211].

Finally, rTMS provides the additional purpose of anticipating the individual responsiveness of patients who may undergo subsequent implantation of a device for MCS [212]. In fact, a preoperative trial utilizing rTMS has demonstrated the ability to predict satisfactory pain relief in approximately 80 to 90% of cases where MCS implantation is performed [213, 214].

Transcutaneous electrical nerve stimulation

Transcutaneous Electrical Nerve Stimulation (TENS) is a non-invasive therapeutic technique based on the application of low-voltage electrical currents to the skin surface, through electrodes placed on or near the area of pain or along the nerve pathways [215]. The principles of TENS stay in the Gate Control Theory (see above) [7].

TENS is commonly employed for various types of pain. TENS has been studied to treat chronic low back pain. Two large randomized controlled trials were firstly conducted with contrasting results, mainly due to the different design, population, and modulation settings [216, 217]. A very recent systematic review, including the whole literature on this topic (i.e. 17 randomized controlled trials with a total of 1027 adults), has suggested that TENS may marginally reduce the perceived chronic low back pain for a short period, to an extent that cannot be considered clinically relevant [218]. TENS has also been tested in 43 patients with fibromyalgia, demonstrating that one 30-min application improved the pain perception during movement, but not at rest [219]. Then, several studies have been conducted so far in patients with fibromyalgia; the most recent pooled data analysis suggests that TENS improves pain symptoms in this population of patients [220]. A modified system for TENS specifically developed to stimulate the terminal supraorbital branch of the trigeminal nerve has been also demonstrated to prevent [221] or to treat attacks of episodic migraine [222]. However, the current evidence is still weak to recommend its use in this scenario.

Transcranial focused ultrasound

Transcranial Focused Ultrasound (tFUS) for chronic pain represents an emerging non-invasive neurostimulation method that utilizes focused ultrasound waves to modulate brain activity for therapeutic purposes [223]. This technique is characterized by a high spatial resolution, targeting any site of the peripheral or central nervous system [223].

tFUS induces mechanical vibrations and thermal effects influencing the neurons excitability, altering the synaptic transmission, and affecting the excitatory/inhibitory balance of neurotransmitters [224]. tFUS may also release endorphins or other neurochemicals involved in pain modulation [224]. In the application for chronic pain treatment, tFUS is centered to specific areas involved in the pain perception, such as the somatosensory cortex, thalamus, or limbic system [225,226,227].

Today, tFUS is approved in Europe for neuromodulation in neuropathic chronic pain not responsive to conventional pharmacological treatments [228]. Indeed, data available are limited to chronic neuropathic pain, phantom limb pain, neuropathic pain related to spinal cord injury in clinical and preclinical settings [229,230,231].

Future perspectives of neurostimulation

Although the growing literature investigating the complexity of pain transmission and the biochemical effects of neurostimulation, clear indications and strong recommendations still lack. The complexity of anatomical and physiological pathways of pain modulation, together with the various etiopathogenesis and pathophysiology of different forms of chronic pain, should probably require a personalized approach to neurostimulation.

Patients can develop chronic pain through various pathophysiological pathways. However, any single treatment, whether pharmacological or involving neurostimulation, has a finite number of mechanisms of action. Consequently, it may only be effective for a specific subset of patients. Monotherapy, whether it's pharmacological, neurostimulatory, surgical, or psychological, is thus likely to result in some individuals who do not respond adequately. These individuals, termed "non-responders," have underlying disease mechanisms that do not align with the mechanism of action of the chosen therapy [212, 232].

Novel approaches to neurostimulation, inspired by the principles of the personalized and precision medicine, have been proposed for other diseases, like tinnitus [233,234,235,236,237], Parkinson’s disease [238, 239], epilepsy [240] and depression [241, 242]. A recent publication explores the reasoning and basis for personalized electrical neurostimulation in individuals suffering from chronic neuropathic pain [212].Noninvasive methods of neurostimulation may predict the response of every single patient to more invasive techniques such a MCS or DBS [212]. Beside the prediction of responsiveness to a specific treatment, the study of clinical phenotypes [243], brain networks and oscillatory patterns [244,245,246,247], and patient’s genotype with the corresponding single nucleotide polymorphism [248,249,250,251] may also guide the clinician in the adoption of the best strategy for every single patient. In addition, the differences in neuropathophysiological processes underlying various painful syndromes should be taken into consideration when selecting a neurostimulation strategy, as recently suggested [252].

It should also be considered that technical advances (such as microelectronics, feedback-based system design, biomimetic stimulation patterns), and the integration of genomics with device-based therapies are revolutionizing the conceptualization of neuromodulation, with a trend towards miniaturization and noninvasive therapies [253].

In this complex scenario, we believe that the scientific literature should be directed on the way to better clarify those underlying anatomical and pathophysiological mechanisms that are still obscure. Once these bases are clearer, more detailed, and better focused studies could be specifically designed to assess and to define the criteria of choice of one technique over another one, for every single chronic pain manifestation.

Meanwhile, the approach to a patient suffering from chronic pain necessitating neurostimulation demands meticulous evaluation, taking into account the underlying condition, individual characteristics (including psychological factors), and prior treatment modalities. Given the absence of comprehensive cost-effectiveness studies, except for SCS, physicians may opt to transition from non-invasive to more invasive techniques known to be efficacious for that particular painful condition.

Conclusions

The electrical neuromodulation of pain comprises various techniques targeting distinct neural pathways involved in pain modulation. Despite numerous studies and trials, determining the most effective technique for each patient remains challenging. Patient selection plays a pivotal role in achieving optimal outcomes, considering factors such as the specific type and origin of chronic pain. Additionally, the invasiveness of the chosen treatment should align and be proportional with the patient's clinical status. Further trials are essential to refine these considerations. Meanwhile, a personalized approach to neurostimulation should guide clinicians in selecting the most appropriate treatment for individual patients.

Availability of data and materials

No datasets were generated or analysed during the current study.

Abbreviations

5-HT:

5-Hydroxytryptamine

AMPA:

Amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid

CGRP:

Calcitonin Gene-Related Peptide

CNS:

Central Nervous System

DBS:

Deep Brain Stimulation

DRG:

Dorsal Root Ganglion

GABA:

Gamma-Aminobutyric Acid

IASP:

International Association for the Study of Pain

MCS:

Motor Cortex Stimulation

NMDA:

N-Methyl-D-Aspartate

PAG:

PeriAqueductal Grey

PENS:

Percutaneous Electrical Nerve Stimulation

PNS:

Peripheral Nerve Stimulation

PRF:

Pulsed Radiofrequency Therapy

rTMS:

Repetitive Transcranial Magnetic Stimulation

RVM:

RostroVentral Medulla

SCS:

Spinal Cord Stimulation

tDCS:

Transcranial Direct Current Stimulation

TENS:

Transcutaneous Electrical Nerve Stimulation

tFUS:

Transcranial Focused Ultrasound

VNS:

Vagus Nerve Stimulation

References

  1. Raja SN, Carr DB, Cohen M, Finnerup NB, Flor H, Gibson S, Keefe FJ, Mogil JS, Ringkamp M, Sluka KA, Song XJ, Stevens B, Sullivan MD, Tutelman PR, Ushida T, Vader K (2020) The revised International Association for the Study of Pain definition of pain: concepts, challenges, and compromises. Pain 161(9):1976–1982. https://doi.org/10.1097/j.pain.0000000000001939

    Article  PubMed  PubMed Central  Google Scholar 

  2. Nugraha B, Gutenbrunner C, Barke A, Karst M, Schiller J, Schafer P, Falter S, Korwisi B, Rief W, Treede RD (2019) The IASP classification of chronic pain for ICD-11: functioning properties of chronic pain. Pain 160(1):88–94. https://doi.org/10.1097/j.pain.0000000000001433

    Article  PubMed  Google Scholar 

  3. Treede RD, Rief W, Barke A, Aziz Q, Bennett MI, Benoliel R, Cohen M, Evers S, Finnerup NB, First MB, Giamberardino MA, Kaasa S, Korwisi B, Kosek E, Lavand’homme P, Nicholas M, Perrot S, Scholz J, Schug S, Smith BH, Svensson P, Vlaeyen JWS, Wang SJ (2019) Chronic pain as a symptom or a disease: the IASP Classification of Chronic Pain for the International Classification of Diseases (ICD-11). Pain 160(1):19–27. https://doi.org/10.1097/j.pain.0000000000001384

    Article  PubMed  Google Scholar 

  4. Schug SA, Lavand’homme P, Barke A, Korwisi B, Rief W, Treede RD (2019) The IASP classification of chronic pain for ICD-11: chronic postsurgical or posttraumatic pain. Pain 160(1):45–52. https://doi.org/10.1097/j.pain.0000000000001413

    Article  PubMed  Google Scholar 

  5. Fayaz A, Croft P, Langford RM, Donaldson LJ, Jones GT (2016) Prevalence of chronic pain in the UK: a systematic review and meta-analysis of population studies. BMJ Open 6(6):e010364. https://doi.org/10.1136/bmjopen-2015-010364

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Vos T, Flaxman AD, Naghavi M, Lozano R, Michaud C, Ezzati M, Shibuya K, Salomon JA, Abdalla S, Aboyans V, Abraham J, Ackerman I, Aggarwal R, Ahn SY, Ali MK, Alvarado M, Anderson HR, Anderson LM, Andrews KG, Atkinson C, Baddour LM, Bahalim AN, Barker-Collo S, Barrero LH, Bartels DH, Basanez MG, Baxter A, Bell ML, Benjamin EJ, Bennett D, Bernabe E, Bhalla K, Bhandari B, Bikbov B, Bin Abdulhak A, Birbeck G, Black JA, Blencowe H, Blore JD, Blyth F, Bolliger I, Bonaventure A, Boufous S, Bourne R, Boussinesq M, Braithwaite T, Brayne C, Bridgett L, Brooker S, Brooks P, Brugha TS, Bryan-Hancock C, Bucello C, Buchbinder R, Buckle G, Budke CM, Burch M, Burney P, Burstein R, Calabria B, Campbell B, Canter CE, Carabin H, Carapetis J, Carmona L, Cella C, Charlson F, Chen H, Cheng AT, Chou D, Chugh SS, Coffeng LE, Colan SD, Colquhoun S, Colson KE, Condon J, Connor MD, Cooper LT, Corriere M, Cortinovis M, de Vaccaro KC, Couser W, Cowie BC, Criqui MH, Cross M, Dabhadkar KC, Dahiya M, Dahodwala N, Damsere-Derry J, Danaei G, Davis A, De Leo D, Degenhardt L, Dellavalle R, Delossantos A, Denenberg J, Derrett S, Des Jarlais DC, Dharmaratne SD, Dherani M, Diaz-Torne C, Dolk H, Dorsey ER, Driscoll T, Duber H, Ebel B, Edmond K, Elbaz A, Ali SE, Erskine H, Erwin PJ, Espindola P, Ewoigbokhan SE, Farzadfar F, Feigin V, Felson DT, Ferrari A, Ferri CP, Fevre EM, Finucane MM, Flaxman S, Flood L, Foreman K, Forouzanfar MH, Fowkes FG, Franklin R, Fransen M, Freeman MK, Gabbe BJ, Gabriel SE, Gakidou E, Ganatra HA, Garcia B, Gaspari F, Gillum RF, Gmel G, Gosselin R, Grainger R, Groeger J, Guillemin F, Gunnell D, Gupta R, Haagsma J, Hagan H, Halasa YA, Hall W, Haring D, Haro JM, Harrison JE, Havmoeller R, Hay RJ, Higashi H, Hill C, Hoen B, Hoffman H, Hotez PJ, Hoy D, Huang JJ, Ibeanusi SE, Jacobsen KH, James SL, Jarvis D, Jasrasaria R, Jayaraman S, Johns N, Jonas JB, Karthikeyan G, Kassebaum N, Kawakami N, Keren A, Khoo JP, King CH, Knowlton LM, Kobusingye O, Koranteng A, Krishnamurthi R, Lalloo R, Laslett LL, Lathlean T, Leasher JL, Lee YY, Leigh J, Lim SS, Limb E, Lin JK, Lipnick M, Lipshultz SE, Liu W, Loane M, Ohno SL, Lyons R, Ma J, Mabweijano J, MacIntyre MF, Malekzadeh R, Mallinger L, Manivannan S, Marcenes W, March L, Margolis DJ, Marks GB, Marks R, Matsumori A, Matzopoulos R, Mayosi BM, McAnulty JH, McDermott MM, McGill N, McGrath J, Medina-Mora ME, Meltzer M, Mensah GA, Merriman TR, Meyer AC, Miglioli V, Miller M, Miller TR, Mitchell PB, Mocumbi AO, Moffitt TE, Mokdad AA, Monasta L, Montico M, Moradi-Lakeh M, Moran A, Morawska L, Mori R, Murdoch ME, Mwaniki MK, Naidoo K, Nair MN, Naldi L, Narayan KM, Nelson PK, Nelson RG, Nevitt MC, Newton CR, Nolte S, Norman P, Norman R, O’Donnell M, O’Hanlon S, Olives C, Omer SB, Ortblad K, Osborne R, Ozgediz D, Page A, Pahari B, Pandian JD, Rivero AP, Patten SB, Pearce N, Padilla RP, Perez-Ruiz F, Perico N, Pesudovs K, Phillips D, Phillips MR, Pierce K, Pion S, Polanczyk GV, Polinder S, Pope CA 3rd, Popova S, Porrini E, Pourmalek F, Prince M, Pullan RL, Ramaiah KD, Ranganathan D, Razavi H, Regan M, Rehm JT, Rein DB, Remuzzi G, Richardson K, Rivara FP, Roberts T, Robinson C, De Leon FR, Ronfani L, Room R, Rosenfeld LC, Rushton L, Sacco RL, Saha S, Sampson U, Sanchez-Riera L, Sanman E, Schwebel DC, Scott JG, Segui-Gomez M, Shahraz S, Shepard DS, Shin H, Shivakoti R, Singh D, Singh GM, Singh JA, Singleton J, Sleet DA, Sliwa K, Smith E, Smith JL, Stapelberg NJ, Steer A, Steiner T, Stolk WA, Stovner LJ, Sudfeld C, Syed S, Tamburlini G, Tavakkoli M, Taylor HR, Taylor JA, Taylor WJ, Thomas B, Thomson WM, Thurston GD, Tleyjeh IM, Tonelli M, Towbin JA, Truelsen T, Tsilimbaris MK, Ubeda C, Undurraga EA, van der Werf MJ, van Os J, Vavilala MS, Venketasubramanian N, Wang M, Wang W, Watt K, Weatherall DJ, Weinstock MA, Weintraub R, Weisskopf MG, Weissman MM, White RA, Whiteford H, Wiersma ST, Wilkinson JD, Williams HC, Williams SR, Witt E, Wolfe F, Woolf AD, Wulf S, Yeh PH, Zaidi AK, Zheng ZJ, Zonies D, Lopez AD, Murray CJ, AlMazroa MA, Memish ZA (2012) Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 380(9859):2163–2196. https://doi.org/10.1016/S0140-6736(12)61729-2

    Article  PubMed  PubMed Central  Google Scholar 

  7. Guzzi G, Della Torre A, La Torre D, Volpentesta G, Stroscio CA, Lavano A, Longhini F (2022) Spinal Cord Stimulation in Chronic Low Back Pain Syndrome: Mechanisms of Modulation, Technical Features and Clinical Application. Healthcare (Basel) 10 (10). https://doi.org/10.3390/healthcare10101953

  8. Pereira A, Herrero-Trujillano M, Vaquero G, Fuentes L, Gonzalez S, Mendiola A, Perez-Medina T (2022) Clinical Management of Chronic Pelvic Pain in Endometriosis Unresponsive to Conventional Therapy. J Pers Med 12 (1). https://doi.org/10.3390/jpm12010101

  9. Pirvulescu I, Biskis A, Candido KD, Knezevic NN (2022) Overcoming clinical challenges of refractory neuropathic pain. Expert Rev Neurother 22(7):595–622. https://doi.org/10.1080/14737175.2022.2105206

    Article  CAS  PubMed  Google Scholar 

  10. Knotkova H, Hamani C, Sivanesan E, Le Beuffe MFE, Moon JY, Cohen SP, Huntoon MA (2021) Neuromodulation for chronic pain. Lancet 397(10289):2111–2124. https://doi.org/10.1016/S0140-6736(21)00794-7

    Article  PubMed  Google Scholar 

  11. Moisset X, Lanteri-Minet M, Fontaine D (2020) Neurostimulation methods in the treatment of chronic pain. J Neural Transm (Vienna) 127(4):673–686. https://doi.org/10.1007/s00702-019-02092-y

    Article  CAS  PubMed  Google Scholar 

  12. Bourne S, Machado AG, Nagel SJ (2014) Basic anatomy and physiology of pain pathways. Neurosurg Clin N Am 25(4):629–638. https://doi.org/10.1016/j.nec.2014.06.001

    Article  PubMed  Google Scholar 

  13. Gonzalez-Hermosillo DC, Gonzalez-Hermosillo LM, Villasenor-Almaraz M, Ballesteros-Herrera D, Moreno-Jimenez S, Corona-Cedillo R, Velasco-Campos F, Carrillo-Ruiz JD, Roldan-Valadez E (2023) Current concepts of pain pathways: a brief review of anatomy, physiology, and medical imaging. Curr Med Imaging. https://doi.org/10.2174/1573405620666230519144112

    Article  PubMed  Google Scholar 

  14. Hodge CJ Jr, Apkarian AV (1990) The spinothalamic tract. Crit Rev Neurobiol 5(4):363–397

    PubMed  Google Scholar 

  15. de Mello Rosa GH, Ullah F, de Paiva YB, da Silva JA, Branco LGS, Corrado AP, Medeiros P, Coimbra NC, Franceschi Biagioni A (2022) Ventrolateral periaqueductal gray matter integrative system of defense and antinociception. Pflugers Arch 474(4):469–480. https://doi.org/10.1007/s00424-022-02672-0

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Jasmin L, Burkey AR, Granato A, Ohara PT (2004) Rostral agranular insular cortex and pain areas of the central nervous system: a tract-tracing study in the rat. J Comp Neurol 468(3):425–440. https://doi.org/10.1002/cne.10978

    Article  PubMed  Google Scholar 

  17. Mokhtar M, Singh P (2024) Neuroanatomy Periaqueductal Gray (NBK554391 [bookaccession])

    Google Scholar 

  18. Comitato A, Bardoni R (2021) Presynaptic Inhibition of Pain and Touch in the Spinal Cord: From Receptors to Circuits. Int J Mol Sci 22 (1). https://doi.org/10.3390/ijms22010414

  19. Melzack R, Wall PD (1965) Pain mechanisms: a new theory. Science 150(3699):971–979. https://doi.org/10.1126/science.150.3699.971

    Article  CAS  PubMed  Google Scholar 

  20. Ossipov MH (2012) The perception and endogenous modulation of pain. Scientifica (Cairo) 2012:561761. https://doi.org/10.6064/2012/561761

    Article  CAS  PubMed  Google Scholar 

  21. Zieglgansberger W (2019) Substance P and pain chronicity. Cell Tissue Res 375(1):227–241. https://doi.org/10.1007/s00441-018-2922-y

    Article  CAS  PubMed  Google Scholar 

  22. Mantyh PW (2002) Neurobiology of substance P and the NK1 receptor. J Clin Psychiatry 63(Suppl 11):6–10

    CAS  PubMed  Google Scholar 

  23. Graefe SB, Rahimi N, Mohiuddin SS (2024) Biochemistry, Substance P. (NBK554583 [bookaccession])

    Google Scholar 

  24. Yang S, Chang MC (2019) Chronic Pain: Structural and Functional Changes in Brain Structures and Associated Negative Affective States. Int J Mol Sci 20 (13). https://doi.org/10.3390/ijms20133130

  25. Zhou Y, Danbolt NC (2014) Glutamate as a neurotransmitter in the healthy brain. J Neural Transm (Vienna) 121(8):799–817. https://doi.org/10.1007/s00702-014-1180-8

    Article  CAS  PubMed  Google Scholar 

  26. D’Mello R, Dickenson AH (2008) Spinal cord mechanisms of pain. Br J Anaesth 101(1):8–16. https://doi.org/10.1093/bja/aen088

    Article  CAS  PubMed  Google Scholar 

  27. Dedek A, Hildebrand ME (2022) Advances and Barriers in Understanding Presynaptic N-Methyl-D-Aspartate Receptors in Spinal Pain Processing. Front Mol Neurosci 15:864502. https://doi.org/10.3389/fnmol.2022.864502

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Granstein RD, Wagner JA, Stohl LL, Ding W (2015) Calcitonin gene-related peptide: key regulator of cutaneous immunity. Acta Physiol (Oxf) 213(3):586–594. https://doi.org/10.1111/apha.12442

    Article  CAS  PubMed  Google Scholar 

  29. Basbaum AI, Bautista DM, Scherrer G, Julius D (2009) Cellular and molecular mechanisms of pain. Cell 139(2):267–284. https://doi.org/10.1016/j.cell.2009.09.028

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wattiez AS, Sowers LP, Russo AF (2020) Calcitonin gene-related peptide (CGRP): role in migraine pathophysiology and therapeutic targeting. Expert Opin Ther Targets 24(2):91–100. https://doi.org/10.1080/14728222.2020.1724285

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Pertovaara A (2006) Noradrenergic pain modulation. Prog Neurobiol 80(2):53–83. https://doi.org/10.1016/j.pneurobio.2006.08.001

    Article  CAS  PubMed  Google Scholar 

  32. Summers RJ, McMartin LR (1993) Adrenoceptors and their second messenger systems. J Neurochem 60(1):10–23. https://doi.org/10.1111/j.1471-4159.1993.tb05817.x

    Article  CAS  PubMed  Google Scholar 

  33. Aimone LD, Jones SL, Gebhart GF (1987) Stimulation-produced descending inhibition from the periaqueductal gray and nucleus raphe magnus in the rat: mediation by spinal monoamines but not opioids. Pain 31(1):123–136. https://doi.org/10.1016/0304-3959(87)90012-1

    Article  CAS  PubMed  Google Scholar 

  34. Bardin L (2011) The complex role of serotonin and 5-HT receptors in chronic pain. Behav Pharmacol 22(5–6):390–404. https://doi.org/10.1097/FBP.0b013e328349aae4

    Article  CAS  PubMed  Google Scholar 

  35. Bannister K, Dickenson AH (2017) The plasticity of descending controls in pain: translational probing. J Physiol 595(13):4159–4166. https://doi.org/10.1113/JP274165

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Charnay Y, Leger L (2010) Brain serotonergic circuitries. Dialogues Clin Neurosci 12(4):471–487

    Article  PubMed  PubMed Central  Google Scholar 

  37. de Leon AS, Tadi P (2024) Biochemistry Gamma Aminobutyric Acid (NBK551683 [bookaccession])

    Google Scholar 

  38. Zeilhofer HU, Wildner H, Yevenes GE (2012) Fast synaptic inhibition in spinal sensory processing and pain control. Physiol Rev 92(1):193–235. https://doi.org/10.1152/physrev.00043.2010

    Article  CAS  PubMed  Google Scholar 

  39. Francois A, Low SA, Sypek EI, Christensen AJ, Sotoudeh C, Beier KT, Ramakrishnan C, Ritola KD, Sharif-Naeini R, Deisseroth K, Delp SL, Malenka RC, Luo L, Hantman AW, Scherrer G (2017) A Brainstem-Spinal Cord Inhibitory Circuit for Mechanical Pain Modulation by GABA and Enkephalins. Neuron 93(4):822-839.e826. https://doi.org/10.1016/j.neuron.2017.01.008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Pathan H, Williams J (2012) Basic opioid pharmacology: an update. Br J Pain 6(1):11–16. https://doi.org/10.1177/2049463712438493

    Article  PubMed  PubMed Central  Google Scholar 

  41. Snyder SH, Pasternak GW (2003) Historical review: Opioid receptors. Trends Pharmacol Sci 24(4):198–205. https://doi.org/10.1016/S0165-6147(03)00066-X

    Article  CAS  PubMed  Google Scholar 

  42. Yam MF, Loh YC, Tan CS, Khadijah Adam S, Abdul Manan N, Basir R (2018) General Pathways of Pain Sensation and the Major Neurotransmitters Involved in Pain Regulation. Int J Mol Sci 19 (8). https://doi.org/10.3390/ijms19082164

  43. Parraga JP, Castellanos A (2023) A Manifesto in Defense of Pain Complexity: A Critical Review of Essential Insights in Pain Neuroscience. J Clin Med 12 (22). https://doi.org/10.3390/jcm12227080

  44. Gangadharan V, Kuner R (2013) Pain hypersensitivity mechanisms at a glance. Dis Model Mech 6(4):889–895. https://doi.org/10.1242/dmm.011502

    Article  PubMed  PubMed Central  Google Scholar 

  45. Gangadharan V, Wang R, Ulzhofer B, Luo C, Bardoni R, Bali KK, Agarwal N, Tegeder I, Hildebrandt U, Nagy GG, Todd AJ, Ghirri A, Haussler A, Sprengel R, Seeburg PH, MacDermott AB, Lewin GR, Kuner R (2011) Peripheral calcium-permeable AMPA receptors regulate chronic inflammatory pain in mice. J Clin Invest 121(4):1608–1623. https://doi.org/10.1172/JCI44911

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Gold MS, Gebhart GF (2010) Nociceptor sensitization in pain pathogenesis. Nat Med 16(11):1248–1257. https://doi.org/10.1038/nm.2235

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Schweizerhof M, Stosser S, Kurejova M, Njoo C, Gangadharan V, Agarwal N, Schmelz M, Bali KK, Michalski CW, Brugger S, Dickenson A, Simone DA, Kuner R (2009) Hematopoietic colony-stimulating factors mediate tumor-nerve interactions and bone cancer pain. Nat Med 15(7):802–807. https://doi.org/10.1038/nm.1976

    Article  CAS  PubMed  Google Scholar 

  48. Binshtok AM, Wang H, Zimmermann K, Amaya F, Vardeh D, Shi L, Brenner GJ, Ji RR, Bean BP, Woolf CJ, Samad TA (2008) Nociceptors are interleukin-1beta sensors. J Neurosci 28(52):14062–14073. https://doi.org/10.1523/JNEUROSCI.3795-08.2008

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Julius D, Basbaum AI (2001) Molecular mechanisms of nociception. Nature 413(6852):203–210. https://doi.org/10.1038/35093019

    Article  CAS  PubMed  Google Scholar 

  50. Woolf CJ (2011) Central sensitization: implications for the diagnosis and treatment of pain. Pain 152(3 Suppl):S2–S15. https://doi.org/10.1016/j.pain.2010.09.030

    Article  PubMed  Google Scholar 

  51. Woolf CJ (1983) Evidence for a central component of post-injury pain hypersensitivity. Nature 306(5944):686–688. https://doi.org/10.1038/306686a0

    Article  CAS  PubMed  Google Scholar 

  52. Bliss TV, Lomo T (1973) Long-lasting potentiation of synaptic transmission in the dentate area of the anaesthetized rabbit following stimulation of the perforant path. J Physiol 232(2):331–356. https://doi.org/10.1113/jphysiol.1973.sp010273

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Nijs J, George SZ, Clauw DJ, Fernandez-de-Las-Penas C, Kosek E, Ickmans K, Fernandez-Carnero J, Polli A, Kapreli E, Huysmans E, Cuesta-Vargas AI, Mani R, Lundberg M, Leysen L, Rice D, Sterling M, Curatolo M (2021) Central sensitisation in chronic pain conditions: latest discoveries and their potential for precision medicine. Lancet Rheumatol 3(5):e383–e392. https://doi.org/10.1016/S2665-9913(21)00032-1

    Article  CAS  PubMed  Google Scholar 

  54. Yu K, Niu X, He B (2020) Neuromodulation Management of Chronic Neuropathic Pain in The Central Nervous system. Adv Funct Mater 30 (37). https://doi.org/10.1002/adfm.201908999

  55. Li SL, Li J, Xu HC, Liu YC, Yang TT, Yuan H (2022) Progress in the efficacy and mechanism of spinal cord stimulation in neuropathological pain. Ibrain 8(1):23–36. https://doi.org/10.1002/ibra.12020

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Deer TR, Eldabe S, Falowski SM, Huntoon MA, Staats PS, Cassar IR, Crosby ND, Boggs JW (2021) Peripherally Induced Reconditioning of the Central Nervous System: A Proposed Mechanistic Theory for Sustained Relief of Chronic Pain with Percutaneous Peripheral Nerve Stimulation. J Pain Res 14:721–736. https://doi.org/10.2147/JPR.S297091

    Article  PubMed  PubMed Central  Google Scholar 

  57. Giannoni-Luza S, Pacheco-Barrios K, Cardenas-Rojas A, Mejia-Pando PF, Luna-Cuadros MA, Barouh JL, Gnoatto-Medeiros M, Candido-Santos L, Barra A, Caumo W, Fregni F (2020) Noninvasive motor cortex stimulation effects on quantitative sensory testing in healthy and chronic pain subjects: a systematic review and meta-analysis. Pain 161(9):1955–1975. https://doi.org/10.1097/j.pain.0000000000001893

    Article  PubMed  PubMed Central  Google Scholar 

  58. Lopes PSS, Campos ACP, Fonoff ET, Britto LRG, Pagano RL (2019) Motor cortex and pain control: exploring the descending relay analgesic pathways and spinal nociceptive neurons in healthy conscious rats. Behav Brain Funct 15(1):5. https://doi.org/10.1186/s12993-019-0156-0

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Klein MM, Treister R, Raij T, Pascual-Leone A, Park L, Nurmikko T, Lenz F, Lefaucheur JP, Lang M, Hallett M, Fox M, Cudkowicz M, Costello A, Carr DB, Ayache SS, Oaklander AL (2015) Transcranial magnetic stimulation of the brain: guidelines for pain treatment research. Pain 156(9):1601–1614. https://doi.org/10.1097/j.pain.0000000000000210

    Article  PubMed  PubMed Central  Google Scholar 

  60. Mariano TY, Burgess FW, Bowker M, Kirschner J, Van’t Wout-Frank M, Jones RN, Halladay CW, Stein M, Greenberg BD (2019) Transcranial Direct Current Stimulation for Affective Symptoms and Functioning in Chronic Low Back Pain: A Pilot Double-Blinded, Randomized. Placebo-Controlled Trial Pain Med 20(6):1166–1177. https://doi.org/10.1093/pm/pny188

    Article  PubMed  Google Scholar 

  61. Machado AG, Baker KB, Plow E, Malone DA (2013) Cerebral stimulation for the affective component of neuropathic pain. Neuromodulation 16(6):514–518. https://doi.org/10.1111/j.1525-1403.2012.00517.x

    Article  PubMed  Google Scholar 

  62. Tsubokawa T, Katayama Y, Yamamoto T, Hirayama T, Koyama S (1993) Chronic motor cortex stimulation in patients with thalamic pain. J Neurosurg 78(3):393–401. https://doi.org/10.3171/jns.1993.78.3.0393

    Article  CAS  PubMed  Google Scholar 

  63. Tsubokawa T, Katayama Y, Yamamoto T, Hirayama T, Koyama S (1991) Treatment of thalamic pain by chronic motor cortex stimulation. Pacing Clin Electrophysiol 14(1):131–134. https://doi.org/10.1111/j.1540-8159.1991.tb04058.x

    Article  CAS  PubMed  Google Scholar 

  64. Lavano A, Guzzi G, M DER, Romano M, Della Torre A, Vescio G, Deodato F, Lavano F, Volpentesta G, (2017) Minimally invasive motor cortex stimulation for Parkinson’s disease. J Neurosurg Sci 61(1):77–87. https://doi.org/10.23736/S0390-5616.16.03246-X

    Article  PubMed  Google Scholar 

  65. Tronnier V, Rasche D (2013) Epidural and subdural stimulation. Handb Clin Neurol 116:343–351. https://doi.org/10.1016/B978-0-444-53497-2.00028-0

    Article  CAS  PubMed  Google Scholar 

  66. Zhang X, Hu Y, Tao W, Zhu H, Xiao D, Li Y (2017) The Effect of Motor Cortex Stimulation on Central Poststroke Pain in a Series of 16 Patients With a Mean Follow-Up of 28 Months. Neuromodulation 20(5):492–496. https://doi.org/10.1111/ner.12547

    Article  PubMed  Google Scholar 

  67. Garcia-Larrea L, Peyron R (2007) Motor cortex stimulation for neuropathic pain: From phenomenology to mechanisms. Neuroimage 37(Suppl 1):S71-79. https://doi.org/10.1016/j.neuroimage.2007.05.062

    Article  PubMed  Google Scholar 

  68. Ramos-Fresnedo A, Perez-Vega C, Domingo RA, Cheshire WP, Middlebrooks EH, Grewal SS (2022) Motor Cortex Stimulation for Pain: A Narrative Review of Indications, Techniques, and Outcomes. Neuromodulation 25(2):211–221. https://doi.org/10.1016/j.neurom.2021.10.025

    Article  PubMed  Google Scholar 

  69. Hamani C, Fonoff ET, Parravano DC, Silva VA, Galhardoni R, Monaco BA, Navarro J, Yeng LT, Teixeira MJ, de Andrade DC (2021) Motor cortex stimulation for chronic neuropathic pain: results of a double-blind randomized study. Brain 144(10):2994–3004. https://doi.org/10.1093/brain/awab189

    Article  PubMed  Google Scholar 

  70. Henssen D, Kurt E, van Walsum AVC, Kozicz T, van Dongen R, Bartels R (2020) Motor cortex stimulation in chronic neuropathic orofacial pain syndromes: a systematic review and meta-analysis. Sci Rep 10(1):7195. https://doi.org/10.1038/s41598-020-64177-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Parravano DC, Ciampi DA, Fonoff ET, Monaco B, Navarro J, Yeng LT, Teixeira MJ, Hamani C (2019) Quality of Life After Motor Cortex Stimulation: Clinical Results and Systematic Review of the Literature. Neurosurgery 84(2):451–456. https://doi.org/10.1093/neuros/nyy060

    Article  PubMed  Google Scholar 

  72. Lavano A, Guzzi G, Chirchiglia D (2018) Cortical neuromodulation for neuropathic pain and Parkinson disease: Where are we? Neurol Neurochir Pol 52(1):75–78. https://doi.org/10.1016/j.pjnns.2017.11.001

    Article  PubMed  Google Scholar 

  73. Rasche D, Tronnier VM (2016) Clinical Significance of Invasive Motor Cortex Stimulation for Trigeminal Facial Neuropathic Pain Syndromes. Neurosurgery 79(5):655–666. https://doi.org/10.1227/NEU.0000000000001353

    Article  PubMed  Google Scholar 

  74. Sokal P, Harat M, Zielinski P, Furtak J, Paczkowski D, Rusinek M (2015) Motor cortex stimulation in patients with chronic central pain. Adv Clin Exp Med 24(2):289–296. https://doi.org/10.17219/acem/40452

    Article  PubMed  Google Scholar 

  75. Szymoniuk M, Chin JH, Domagalski L, Biszewski M, Jozwik K, Kamieniak P (2023) Brain stimulation for chronic pain management: a narrative review of analgesic mechanisms and clinical evidence. Neurosurg Rev 46(1):127. https://doi.org/10.1007/s10143-023-02032-1

    Article  PubMed  PubMed Central  Google Scholar 

  76. Bergeron D, Obaid S, Fournier-Gosselin MP, Bouthillier A, Nguyen DK (2021) Deep Brain Stimulation of the Posterior Insula in Chronic Pain: A Theoretical Framework. Brain Sci 11 (5). https://doi.org/10.3390/brainsci11050639

  77. Mancini F, Guzzi G, Castrioto CF, Cavallo MA, Conti A, Conti C, Della Torre A, Esposito F, Iorio G, Landi A, Lanotte M, Lavano A, Locatelli M, Longhi M, Marruzzo D, Mondani M, Morace R, Pellizzari M, Piacentino M, Picozzi P, Romeo F, Sarrubbo S, Servello D, Somma T, Trezza A, Tringali G, Tufo T, Ricciuti RA (2022) Deep brain stimulation for Parkinson’s disease in practice: results of the survey by the Italian Neurosurgery Society. J Neurosurg Sci 66(6):526–534. https://doi.org/10.23736/S0390-5616.22.05751-4

    Article  PubMed  Google Scholar 

  78. Sims-Williams H, Matthews JC, Talbot PS, Love-Jones S, Brooks JC, Patel NK, Pickering AE (2017) Deep brain stimulation of the periaqueductal gray releases endogenous opioids in humans. Neuroimage 146:833–842. https://doi.org/10.1016/j.neuroimage.2016.08.038

    Article  CAS  PubMed  Google Scholar 

  79. Young RF, Bach FW, Van Norman AS, Yaksh TL (1993) Release of beta-endorphin and methionine-enkephalin into cerebrospinal fluid during deep brain stimulation for chronic pain. Effects of stimulation locus and site of sampling. J Neurosurg. 79(6):816–825. https://doi.org/10.3171/jns.1993.79.6.0816

    Article  CAS  PubMed  Google Scholar 

  80. Gopalakrishnan R, Burgess RC, Malone DA, Lempka SF, Gale JT, Floden DP, Baker KB, Machado AG (2018) Deep brain stimulation of the ventral striatal area for poststroke pain syndrome: a magnetoencephalography study. J Neurophysiol 119(6):2118–2128. https://doi.org/10.1152/jn.00830.2017

    Article  PubMed  PubMed Central  Google Scholar 

  81. Lempka SF, Malone DA Jr, Hu B, Baker KB, Wyant A, Ozinga JGt, Plow EB, Pandya M, Kubu CS, Ford PJ, Machado AG, (2017) Randomized clinical trial of deep brain stimulation for poststroke pain. Ann Neurol 81(5):653–663. https://doi.org/10.1002/ana.24927

    Article  PubMed  Google Scholar 

  82. Belasen A, Rizvi K, Gee LE, Yeung P, Prusik J, Ramirez-Zamora A, Hanspal E, Paiva P, Durphy J, Argoff CE, Pilitsis JG (2017) Effect of low-frequency deep brain stimulation on sensory thresholds in Parkinson’s disease. J Neurosurg 126(2):397–403. https://doi.org/10.3171/2016.2.JNS152231

    Article  PubMed  Google Scholar 

  83. Marques A, Chassin O, Morand D, Pereira B, Debilly B, Derost P, Ulla M, Lemaire JJ, Durif F (2013) Central pain modulation after subthalamic nucleus stimulation: A crossover randomized trial. Neurology 81(7):633–640. https://doi.org/10.1212/WNL.0b013e3182a08d00

    Article  PubMed  Google Scholar 

  84. Son BC, Kim DR, Kim HS, Lee SW (2014) Simultaneous trial of deep brain and motor cortex stimulation in chronic intractable neuropathic pain. Stereotact Funct Neurosurg 92(4):218–226. https://doi.org/10.1159/000362933

    Article  PubMed  Google Scholar 

  85. Rasche D, Rinaldi PC, Young RF, Tronnier VM (2006) Deep brain stimulation for the treatment of various chronic pain syndromes. Neurosurg Focus 21(6):E8. https://doi.org/10.3171/foc.2006.21.6.10

    Article  PubMed  Google Scholar 

  86. Yamamoto T, Katayama Y, Obuchi T, Kano T, Kobayashi K, Oshima H, Fukaya C (2006) Thalamic sensory relay nucleus stimulation for the treatment of peripheral deafferentation pain. Stereotact Funct Neurosurg 84(4):180–183. https://doi.org/10.1159/000094958

    Article  PubMed  Google Scholar 

  87. Cruccu G, Garcia-Larrea L, Hansson P, Keindl M, Lefaucheur JP, Paulus W, Taylor R, Tronnier V, Truini A, Attal N (2016) EAN guidelines on central neurostimulation therapy in chronic pain conditions. Eur J Neurol 23(10):1489–1499. https://doi.org/10.1111/ene.13103

    Article  CAS  PubMed  Google Scholar 

  88. Gouveia FV, Warsi NM, Suresh H, Matin R, Ibrahim GM (2024) Neurostimulation treatments for epilepsy: Deep brain stimulation, responsive neurostimulation and vagus nerve stimulation. Neurotherapeutics 21(3):e00308. https://doi.org/10.1016/j.neurot.2023.e00308

    Article  PubMed  Google Scholar 

  89. Jung B, Yang C, Lee SH (2024) Vagus Nerves Stimulation: Clinical Implication and Practical Issue as a Neuropsychiatric Treatment. Clin Psychopharmacol Neurosci 22(1):13–22. https://doi.org/10.9758/cpn.23.1101

    Article  PubMed  Google Scholar 

  90. Shao P, Li H, Jiang J, Guan Y, Chen X, Wang Y (2023) Role of Vagus Nerve Stimulation in the Treatment of Chronic Pain. NeuroImmunoModulation 30(1):167–183. https://doi.org/10.1159/000531626

    Article  CAS  PubMed  Google Scholar 

  91. Chakravarthy K, Chaudhry H, Williams K, Christo PJ (2015) Review of the Uses of Vagal Nerve Stimulation in Chronic Pain Management. Curr Pain Headache Rep 19(12):54. https://doi.org/10.1007/s11916-015-0528-6

    Article  PubMed  Google Scholar 

  92. Cechetto DF (1987) Central representation of visceral function. Fed Proc 46(1):17–23

    CAS  PubMed  Google Scholar 

  93. Ricardo JA, Koh ET (1978) Anatomical evidence of direct projections from the nucleus of the solitary tract to the hypothalamus, amygdala, and other forebrain structures in the rat. Brain Res 153(1):1–26. https://doi.org/10.1016/0006-8993(78)91125-3

    Article  CAS  PubMed  Google Scholar 

  94. Morest DK (1967) Experimental study of the projections of the nucleus of the tractus solitarius and the area postrema in the cat. J Comp Neurol 130(4):277–300. https://doi.org/10.1002/cne.901300402

    Article  CAS  PubMed  Google Scholar 

  95. Vida G, Pena G, Deitch EA, Ulloa L (2011) alpha7-cholinergic receptor mediates vagal induction of splenic norepinephrine. J Immunol 186(7):4340–4346. https://doi.org/10.4049/jimmunol.1003722

    Article  CAS  PubMed  Google Scholar 

  96. Shytle RD, Mori T, Townsend K, Vendrame M, Sun N, Zeng J, Ehrhart J, Silver AA, Sanberg PR, Tan J (2004) Cholinergic modulation of microglial activation by alpha 7 nicotinic receptors. J Neurochem 89(2):337–343. https://doi.org/10.1046/j.1471-4159.2004.02347.x

    Article  CAS  PubMed  Google Scholar 

  97. Hosoi T, Okuma Y, Nomura Y (2000) Electrical stimulation of afferent vagus nerve induces IL-1beta expression in the brain and activates HPA axis. Am J Physiol Regul Integr Comp Physiol 279(1):R141-147. https://doi.org/10.1152/ajpregu.2000.279.1.R141

    Article  CAS  PubMed  Google Scholar 

  98. Tao X, Lee MS, Donnelly CR, Ji RR (2020) Neuromodulation, Specialized Proresolving Mediators, and Resolution of Pain. Neurotherapeutics 17(3):886–899. https://doi.org/10.1007/s13311-020-00892-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Chaudhry SR, Lendvai IS, Muhammad S, Westhofen P, Kruppenbacher J, Scheef L, Boecker H, Scheele D, Hurlemann R, Kinfe TM (2019) Inter-ictal assay of peripheral circulating inflammatory mediators in migraine patients under adjunctive cervical non-invasive vagus nerve stimulation (nVNS): A proof-of-concept study. Brain Stimul 12(3):643–651. https://doi.org/10.1016/j.brs.2019.01.008

    Article  PubMed  Google Scholar 

  100. Lerman I, Hauger R, Sorkin L, Proudfoot J, Davis B, Huang A, Lam K, Simon B, Baker DG (2016) Noninvasive Transcutaneous Vagus Nerve Stimulation Decreases Whole Blood Culture-Derived Cytokines and Chemokines: A Randomized, Blinded. Healthy Control Pilot Trial Neuromodulation 19(3):283–290. https://doi.org/10.1111/ner.12398

    Article  PubMed  Google Scholar 

  101. Tal M (1999) A Role for Inflammation in Chronic Pain. Curr Rev Pain 3(6):440–446. https://doi.org/10.1007/s11916-999-0071-4

    Article  CAS  PubMed  Google Scholar 

  102. Hord ED, Evans MS, Mueed S, Adamolekun B, Naritoku DK (2003) The effect of vagus nerve stimulation on migraines. J Pain 4(9):530–534. https://doi.org/10.1016/j.jpain.2003.08.001

    Article  PubMed  Google Scholar 

  103. Sadler RM, Purdy RA, Rahey S (2002) Vagal nerve stimulation aborts migraine in patient with intractable epilepsy. Cephalalgia 22(6):482–484. https://doi.org/10.1046/j.1468-2982.2002.00387.x

    Article  CAS  PubMed  Google Scholar 

  104. Kirchner A, Birklein F, Stefan H, Handwerker HO (2000) Left vagus nerve stimulation suppresses experimentally induced pain. Neurology 55(8):1167–1171. https://doi.org/10.1212/wnl.55.8.1167

    Article  CAS  PubMed  Google Scholar 

  105. Multon S, Schoenen J (2005) Pain control by vagus nerve stimulation: from animal to man and back. Acta Neurol Belg. 105(2):62–67

    PubMed  Google Scholar 

  106. Najib U, Smith T, Hindiyeh N, Saper J, Nye B, Ashina S, McClure CK, Marmura MJ, Chase S, Liebler E, Lipton RB (2022) Non-invasive vagus nerve stimulation for prevention of migraine: The multicenter, randomized, double-blind, sham-controlled PREMIUM II trial. Cephalalgia 42(7):560–569. https://doi.org/10.1177/03331024211068813

    Article  PubMed  Google Scholar 

  107. Silberstein SD, Calhoun AH, Lipton RB, Grosberg BM, Cady RK, Dorlas S, Simmons KA, Mullin C, Liebler EJ, Goadsby PJ, Saper JR (2016) Chronic migraine headache prevention with noninvasive vagus nerve stimulation: The EVENT study. Neurology 87(5):529–538. https://doi.org/10.1212/WNL.0000000000002918

    Article  PubMed  PubMed Central  Google Scholar 

  108. Barbanti P, Grazzi L, Egeo G, Padovan AM, Liebler E, Bussone G (2015) Non-invasive vagus nerve stimulation for acute treatment of high-frequency and chronic migraine: an open-label study. J Headache Pain 16:61. https://doi.org/10.1186/s10194-015-0542-4

    Article  PubMed  PubMed Central  Google Scholar 

  109. Goadsby PJ, Grosberg BM, Mauskop A, Cady R, Simmons KA (2014) Effect of noninvasive vagus nerve stimulation on acute migraine: an open-label pilot study. Cephalalgia 34(12):986–993. https://doi.org/10.1177/0333102414524494

    Article  CAS  PubMed  Google Scholar 

  110. Lange G, Janal MN, Maniker A, Fitzgibbons J, Fobler M, Cook D, Natelson BH (2011) Safety and efficacy of vagus nerve stimulation in fibromyalgia: a phase I/II proof of concept trial. Pain Med 12(9):1406–1413. https://doi.org/10.1111/j.1526-4637.2011.01203.x

    Article  PubMed  Google Scholar 

  111. Kutlu N, Ozden AV, Alptekin HK, Alptekin JO (2020) The Impact of Auricular Vagus Nerve Stimulation on Pain and Life Quality in Patients with Fibromyalgia Syndrome. Biomed Res Int 2020:8656218. https://doi.org/10.1155/2020/8656218

    Article  PubMed  PubMed Central  Google Scholar 

  112. Wang S, Li S, Zhai X, Rong P, He J, Liu L, He X, Liu W (2022) Transcutaneous auricular vagal nerve stimulation releases extrapineal melatonin and reduces thermal hypersensitivity in Zucker diabetic fatty rats. Front Neurosci 16:916822. https://doi.org/10.3389/fnins.2022.916822

    Article  PubMed  PubMed Central  Google Scholar 

  113. Li S, Sun C, Rong P, Zhai X, Zhang J, Baker M, Wang S (2018) Auricular vagus nerve stimulation enhances central serotonergic function and inhibits diabetic neuropathy development in Zucker fatty rats. Mol Pain 14:1744806918787368. https://doi.org/10.1177/1744806918787368

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Wang S, Zhai X, Li S, McCabe MF, Wang X, Rong P (2015) Transcutaneous vagus nerve stimulation induces tidal melatonin secretion and has an antidiabetic effect in Zucker fatty rats. PLoS ONE 10(4):e0124195. https://doi.org/10.1371/journal.pone.0124195

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Oshinsky ML, Murphy AL, Hekierski H Jr, Cooper M, Simon BJ (2014) Noninvasive vagus nerve stimulation as treatment for trigeminal allodynia. Pain 155(5):1037–1042. https://doi.org/10.1016/j.pain.2014.02.009

    Article  PubMed  PubMed Central  Google Scholar 

  116. Saade NE, Tabet MS, Banna NR, Atweh SF, Jabbur SJ (1985) Inhibition of nociceptive evoked activity in spinal neurons through a dorsal column-brainstem-spinal loop. Brain Res 339(1):115–118. https://doi.org/10.1016/0006-8993(85)90627-4

    Article  CAS  PubMed  Google Scholar 

  117. Bantli H, Bloedel JR, Thienprasit P (1975) Supraspinal interactions resulting from experimental dorsal column stimulation. J Neurosurg 42(3):296–300. https://doi.org/10.3171/jns.1975.42.3.0296

    Article  CAS  PubMed  Google Scholar 

  118. Guan Y (2012) Spinal cord stimulation: neurophysiological and neurochemical mechanisms of action. Curr Pain Headache Rep 16(3):217–225. https://doi.org/10.1007/s11916-012-0260-4

    Article  PubMed  Google Scholar 

  119. Sivanesan E, Maher DP, Raja SN, Linderoth B, Guan Y (2019) Supraspinal Mechanisms of Spinal Cord Stimulation for Modulation of Pain: Five Decades of Research and Prospects for the Future. Anesthesiology 130(4):651–665. https://doi.org/10.1097/ALN.0000000000002353

    Article  PubMed  Google Scholar 

  120. Song Z, Ansah OB, Meyerson BA, Pertovaara A, Linderoth B (2013) Exploration of supraspinal mechanisms in effects of spinal cord stimulation: role of the locus coeruleus. Neuroscience 253:426–434. https://doi.org/10.1016/j.neuroscience.2013.09.006

    Article  CAS  PubMed  Google Scholar 

  121. Saade NE, Atweh SF, Privat A, Jabbur SJ (1999) Inhibitory effects from various types of dorsal column and raphe magnus stimulations on nociceptive withdrawal flexion reflexes. Brain Res 846(1):72–86. https://doi.org/10.1016/s0006-8993(99)02003-x

    Article  CAS  PubMed  Google Scholar 

  122. Song Z, Ansah OB, Meyerson BA, Pertovaara A, Linderoth B (2013) The rostroventromedial medulla is engaged in the effects of spinal cord stimulation in a rodent model of neuropathic pain. Neuroscience 247:134–144. https://doi.org/10.1016/j.neuroscience.2013.05.027

    Article  CAS  PubMed  Google Scholar 

  123. Stiller CO, Linderoth B, O’Connor WT, Franck J, Falkenberg T, Ungerstedt U, Brodin E (1995) Repeated spinal cord stimulation decreases the extracellular level of gamma-aminobutyric acid in the periaqueductal gray matter of freely moving rats. Brain Res 699(2):231–241. https://doi.org/10.1016/0006-8993(95)00911-9

    Article  CAS  PubMed  Google Scholar 

  124. Linderoth B, Gazelius B, Franck J, Brodin E (1992) Dorsal column stimulation induces release of serotonin and substance P in the cat dorsal horn. Neurosurgery 31(2):289–296. https://doi.org/10.1227/00006123-199208000-00014. (discussion 296-287)

    Article  CAS  PubMed  Google Scholar 

  125. Schechtmann G, Song Z, Ultenius C, Meyerson BA, Linderoth B (2008) Cholinergic mechanisms involved in the pain relieving effect of spinal cord stimulation in a model of neuropathy. Pain 139(1):136–145. https://doi.org/10.1016/j.pain.2008.03.023

    Article  CAS  PubMed  Google Scholar 

  126. Song Z, Meyerson BA, Linderoth B (2008) Muscarinic receptor activation potentiates the effect of spinal cord stimulation on pain-related behavior in rats with mononeuropathy. Neurosci Lett 436(1):7–12. https://doi.org/10.1016/j.neulet.2008.02.044

    Article  CAS  PubMed  Google Scholar 

  127. Stiller CO, Cui JG, O’Connor WT, Brodin E, Meyerson BA, Linderoth B (1996) Release of gamma-aminobutyric acid in the dorsal horn and suppression of tactile allodynia by spinal cord stimulation in mononeuropathic rats. Neurosurgery 39(2):367–374. https://doi.org/10.1097/00006123-199608000-00026. (discussion 374-365)

    Article  CAS  PubMed  Google Scholar 

  128. Baba H, Yoshimura M, Nishi S, Shimoji K (1994) Synaptic responses of substantia gelatinosa neurones to dorsal column stimulation in rat spinal cord in vitro. J Physiol 478(Pt 1):87–99. https://doi.org/10.1113/jphysiol.1994.sp020232

    Article  PubMed  PubMed Central  Google Scholar 

  129. Cui JG, O’Connor WT, Ungerstedt U, Linderoth B, Meyerson BA (1997) Spinal cord stimulation attenuates augmented dorsal horn release of excitatory amino acids in mononeuropathy via a GABAergic mechanism. Pain 73(1):87–95. https://doi.org/10.1016/s0304-3959(97)00077-8

    Article  CAS  PubMed  Google Scholar 

  130. Lind G, Schechtmann G, Winter J, Meyerson BA, Linderoth B (2008) Baclofen-enhanced spinal cord stimulation and intrathecal baclofen alone for neuropathic pain: Long-term outcome of a pilot study. Eur J Pain 12(1):132–136. https://doi.org/10.1016/j.ejpain.2007.03.011

    Article  CAS  PubMed  Google Scholar 

  131. Wang YY, Wu SX, Liu XY, Wang W, Li YQ (2003) Effects of c-fos antisense oligodeoxynucleotide on 5-HT-induced upregulation of preprodynorphin, preproenkephalin, and glutamic acid decarboxylase mRNA expression in cultured rat spinal dorsal horn neurons. Biochem Biophys Res Commun 309(3):631–636. https://doi.org/10.1016/j.bbrc.2003.08.046

    Article  CAS  PubMed  Google Scholar 

  132. Song Z, Meyerson BA, Linderoth B (2011) Spinal 5-HT receptors that contribute to the pain-relieving effects of spinal cord stimulation in a rat model of neuropathy. Pain 152(7):1666–1673. https://doi.org/10.1016/j.pain.2011.03.012

    Article  CAS  PubMed  Google Scholar 

  133. Nagel SJ, Lempka SF, Machado AG (2014) Percutaneous spinal cord stimulation for chronic pain: indications and patient selection. Neurosurg Clin N Am 25(4):723–733. https://doi.org/10.1016/j.nec.2014.06.005

    Article  PubMed  Google Scholar 

  134. Sitzman BT, Provenzano DA (2017) Best Practices in Spinal Cord Stimulation. Spine (Phila Pa) 42(Suppl 14):S67–S71. https://doi.org/10.1097/BRS.0000000000002220

    Article  Google Scholar 

  135. Deer TR, Mekhail N, Provenzano D, Pope J, Krames E, Leong M, Levy RM, Abejon D, Buchser E, Burton A, Buvanendran A, Candido K, Caraway D, Cousins M, DeJongste M, Diwan S, Eldabe S, Gatzinsky K, Foreman RD, Hayek S, Kim P, Kinfe T, Kloth D, Kumar K, Rizvi S, Lad SP, Liem L, Linderoth B, Mackey S, McDowell G, McRoberts P, Poree L, Prager J, Raso L, Rauck R, Russo M, Simpson B, Slavin K, Staats P, Stanton-Hicks M, Verrills P, Wellington J, Williams K, North R (2014) The appropriate use of neurostimulation of the spinal cord and peripheral nervous system for the treatment of chronic pain and ischemic diseases: the Neuromodulation Appropriateness Consensus Committee. Neuromodulation 17(6):515–550. https://doi.org/10.1111/ner.12208. (discussion 550)

    Article  PubMed  Google Scholar 

  136. Kumar K, Hunter G, Demeria D (2006) Spinal cord stimulation in treatment of chronic benign pain: challenges in treatment planning and present status, a 22-year experience. Neurosurgery 58(3):481–496. https://doi.org/10.1227/01.NEU.0000192162.99567.96. (discussion 481-496)

    Article  PubMed  Google Scholar 

  137. Kumar K, Rizvi S, Bnurs SB (2011) Spinal cord stimulation is effective in management of complex regional pain syndrome I: fact or fiction. Neurosurgery 69(3):566–578. https://doi.org/10.1227/NEU.0b013e3182181e60. (discussion 5578-5580)

    Article  PubMed  Google Scholar 

  138. Kumar K, Taylor RS, Jacques L, Eldabe S, Meglio M, Molet J, Thomson S, O’Callaghan J, Eisenberg E, Milbouw G, Buchser E, Fortini G, Richardson J, North RB (2008) The effects of spinal cord stimulation in neuropathic pain are sustained: a 24-month follow-up of the prospective randomized controlled multicenter trial of the effectiveness of spinal cord stimulation. Neurosurgery 63(4):762–770. https://doi.org/10.1227/01.NEU.0000325731.46702.D9. (discussion 770)

    Article  PubMed  Google Scholar 

  139. Kumar K, Rizvi S, Nguyen R, Abbas M, Bishop S, Murthy V (2014) Impact of wait times on spinal cord stimulation therapy outcomes. Pain Pract 14(8):709–720. https://doi.org/10.1111/papr.12126

    Article  PubMed  Google Scholar 

  140. Fama CA, Chen N, Prusik J, Kumar V, Wilock M, Roth S, Pilitsis JG (2016) The Use of Preoperative Psychological Evaluations to Predict Spinal Cord Stimulation Success: Our Experience and a Review of the Literature. Neuromodulation 19(4):429–436. https://doi.org/10.1111/ner.12434

    Article  PubMed  Google Scholar 

  141. Grider JS, Manchikanti L, Carayannopoulos A, Sharma ML, Balog CC, Harned ME, Grami V, Justiz R, Nouri KH, Hayek SM, Vallejo R, Christo PJ (2016) Effectiveness of Spinal Cord Stimulation in Chronic Spinal Pain: A Systematic Review. Pain Physician 19(1):E33-54

    Article  PubMed  Google Scholar 

  142. Kapural L, Yu C, Doust MW, Gliner BE, Vallejo R, Sitzman BT, Amirdelfan K, Morgan DM, Brown LL, Yearwood TL, Bundschu R, Burton AW, Yang T, Benyamin R, Burgher AH (2015) Novel 10-kHz High-frequency Therapy (HF10 Therapy) Is Superior to Traditional Low-frequency Spinal Cord Stimulation for the Treatment of Chronic Back and Leg Pain: The SENZA-RCT Randomized Controlled Trial. Anesthesiology 123(4):851–860. https://doi.org/10.1097/ALN.0000000000000774

    Article  PubMed  Google Scholar 

  143. Taylor RS (2006) Spinal cord stimulation in complex regional pain syndrome and refractory neuropathic back and leg pain/failed back surgery syndrome: results of a systematic review and meta-analysis. J Pain Symptom Manage 31(4 Suppl):S13-19. https://doi.org/10.1016/j.jpainsymman.2005.12.010

    Article  PubMed  Google Scholar 

  144. Eldabe S, Thomson S, Duarte R, Brookes M, deBelder M, Raphael J, Davies E, Taylor R (2016) The Effectiveness and Cost-Effectiveness of Spinal Cord Stimulation for Refractory Angina (RASCAL Study): A Pilot Randomized Controlled Trial. Neuromodulation 19(1):60–70. https://doi.org/10.1111/ner.12349

    Article  PubMed  Google Scholar 

  145. Amann W, Berg P, Gersbach P, Gamain J, Raphael JH, Ubbink DT (2003) Spinal cord stimulation in the treatment of non-reconstructable stable critical leg ischaemia: results of the European Peripheral Vascular Disease Outcome Study (SCS-EPOS). Eur J Vasc Endovasc Surg 26(3):280–286. https://doi.org/10.1053/ejvs.2002.1876

    Article  CAS  PubMed  Google Scholar 

  146. Naoum JJ, Arbid EJ (2013) Spinal cord stimulation for chronic limb ischemia. Methodist Debakey Cardiovasc J 9(2):99–102. https://doi.org/10.14797/mdcj-9-2-99

    Article  PubMed  PubMed Central  Google Scholar 

  147. Fontaine R, Kim M, Kieny R (1954) Surgical treatment of peripheral circulation disorders. Helv Chir Acta 21(5–6):499–533

    CAS  PubMed  Google Scholar 

  148. Chapman KB, Sayed D, Lamer T, Hunter C, Weisbein J, Patel KV, Dickerson D, Hagedorn JM, Lee DW, Amirdelfan K, Deer T, Chakravarthy K (2023) Best Practices for Dorsal Root Ganglion Stimulation for Chronic Pain: Guidelines from the American Society of Pain and Neuroscience. J Pain Res 16:839–879. https://doi.org/10.2147/JPR.S364370

    Article  PubMed  PubMed Central  Google Scholar 

  149. Franken G, Douven P, Debets J, Joosten EAJ (2021) Conventional Dorsal Root Ganglion Stimulation in an Experimental Model of Painful Diabetic Peripheral Neuropathy: A Quantitative Immunocytochemical Analysis of Intracellular gamma-Aminobutyric Acid in Dorsal Root Ganglion Neurons. Neuromodulation 24(4):639–645. https://doi.org/10.1111/ner.13398

    Article  PubMed  PubMed Central  Google Scholar 

  150. Koetsier E, Franken G, Debets J, Heijmans L, van Kuijk SMJ, Linderoth B, Joosten EA, Maino P (2020) Mechanism of dorsal root ganglion stimulation for pain relief in painful diabetic polyneuropathy is not dependent on GABA release in the dorsal horn of the spinal cord. CNS Neurosci Ther 26(1):136–143. https://doi.org/10.1111/cns.13192

    Article  CAS  PubMed  Google Scholar 

  151. Chao D, Zhang Z, Mecca CM, Hogan QH, Pan B (2020) Analgesic dorsal root ganglionic field stimulation blocks conduction of afferent impulse trains selectively in nociceptive sensory afferents. Pain 161(12):2872–2886. https://doi.org/10.1097/j.pain.0000000000001982

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Gemes G, Koopmeiners A, Rigaud M, Lirk P, Sapunar D, Bangaru ML, Vilceanu D, Garrison SR, Ljubkovic M, Mueller SJ, Stucky CL, Hogan QH (2013) Failure of action potential propagation in sensory neurons: mechanisms and loss of afferent filtering in C-type units after painful nerve injury. J Physiol 591(4):1111–1131. https://doi.org/10.1113/jphysiol.2012.242750

    Article  CAS  PubMed  Google Scholar 

  153. Devor M (2009) Ectopic discharge in Abeta afferents as a source of neuropathic pain. Exp Brain Res 196(1):115–128. https://doi.org/10.1007/s00221-009-1724-6

    Article  CAS  PubMed  Google Scholar 

  154. Chapman KB, Groenen PS, Vissers KC, van Helmond N, Stanton-Hicks MD (2021) The Pathways and Processes Underlying Spinal Transmission of Low Back Pain: Observations From Dorsal Root Ganglion Stimulation Treatment. Neuromodulation 24(4):610–621. https://doi.org/10.1111/ner.13150

    Article  PubMed  Google Scholar 

  155. Smits H, van Kleef M, Holsheimer J, Joosten EA (2013) Experimental spinal cord stimulation and neuropathic pain: mechanism of action, technical aspects, and effectiveness. Pain Pract 13(2):154–168. https://doi.org/10.1111/j.1533-2500.2012.00579.x

    Article  PubMed  Google Scholar 

  156. Chapman KB, Yousef TA, Foster A, M DS-H, van Helmond N, (2021) Mechanisms for the Clinical Utility of Low-Frequency Stimulation in Neuromodulation of the Dorsal Root Ganglion. Neuromodulation 24(4):738–745. https://doi.org/10.1111/ner.13323

    Article  PubMed  Google Scholar 

  157. Arcourt A, Gorham L, Dhandapani R, Prato V, Taberner FJ, Wende H, Gangadharan V, Birchmeier C, Heppenstall PA, Lechner SG (2017) Touch Receptor-Derived Sensory Information Alleviates Acute Pain Signaling and Fine-Tunes Nociceptive Reflex Coordination. Neuron 93(1):179–193. https://doi.org/10.1016/j.neuron.2016.11.027

    Article  CAS  PubMed  Google Scholar 

  158. Sandkuhler J, Chen JG, Cheng G, Randic M (1997) Low-frequency stimulation of afferent Adelta-fibers induces long-term depression at primary afferent synapses with substantia gelatinosa neurons in the rat. J Neurosci 17(16):6483–6491. https://doi.org/10.1523/JNEUROSCI.17-16-06483.1997

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Chapman KB, Kloosterman J, Schor JA, Girardi GE, van Helmond N, Yousef TA (2021) Objective Improvements in Peripheral Arterial Disease from Dorsal Root Ganglion Stimulation: A Case Series. Ann Vasc Surg 74(519):e517-519e516. https://doi.org/10.1016/j.avsg.2021.01.069

    Article  Google Scholar 

  160. Sverrisdottir YB, Martin SC, Hadjipavlou G, Kent AR, Paterson DJ, FitzGerald JJ, Green AL (2020) Human Dorsal Root Ganglion Stimulation Reduces Sympathetic Outflow and Long-Term Blood Pressure. JACC Basic Transl Sci 5(10):973–985. https://doi.org/10.1016/j.jacbts.2020.07.010

    Article  PubMed  PubMed Central  Google Scholar 

  161. Gravius N, Chaudhry SR, Muhammad S, Bostrom A, Gravius S, Randau T, Scheele D, Westhofen P, Kruppenbacher J, Stoffel-Wagner B, Maier C, Weidlich A, Yearwood TL, Chakravarthy KV, Kramer JM, Hurlemann R, Kinfe TM (2019) Selective L4 Dorsal Root Ganglion Stimulation Evokes Pain Relief and Changes of Inflammatory Markers: Part I Profiling of Saliva and Serum Molecular Patterns. Neuromodulation 22(1):44–52. https://doi.org/10.1111/ner.12866

    Article  PubMed  Google Scholar 

  162. Pan B, Zhang Z, Chao D, Hogan QH (2018) Dorsal Root Ganglion Field Stimulation Prevents Inflammation and Joint Damage in a Rat Model of Rheumatoid Arthritis. Neuromodulation 21(3):247–253. https://doi.org/10.1111/ner.12648

    Article  PubMed  Google Scholar 

  163. Deer TR, Pope JE, Lamer TJ, Grider JS, Provenzano D, Lubenow TR, FitzGerald JJ, Hunter C, Falowski S, Sayed D, Baranidharan G, Patel NK, Davis T, Green A, Pajuelo A, Epstein LJ, Harned M, Liem L, Christo PJ, Chakravarthy K, Gilmore C, Huygen F, Lee E, Metha P, Nijhuis H, Patterson DG, Petersen E, Pilitsis JG, Rowe JJ, Rupert MP, Skaribas I, Sweet J, Verrills P, Wilson D, Levy RM, Mekhail N (2019) The Neuromodulation Appropriateness Consensus Committee on Best Practices for Dorsal Root Ganglion Stimulation. Neuromodulation 22(1):1–35. https://doi.org/10.1111/ner.12845

    Article  PubMed  Google Scholar 

  164. Deer TR, Levy RM, Kramer J, Poree L, Amirdelfan K, Grigsby E, Staats P, Burton AW, Burgher AH, Obray J, Scowcroft J, Golovac S, Kapural L, Paicius R, Kim C, Pope J, Yearwood T, Samuel S, McRoberts WP, Cassim H, Netherton M, Miller N, Schaufele M, Tavel E, Davis T, Davis K, Johnson L, Mekhail N (2017) Dorsal root ganglion stimulation yielded higher treatment success rate for complex regional pain syndrome and causalgia at 3 and 12 months: a randomized comparative trial. Pain 158(4):669–681. https://doi.org/10.1097/j.pain.0000000000000814

    Article  PubMed  Google Scholar 

  165. D’Souza RS, Kubrova E, Her YF, Barman RA, Smith BJ, Alvarez GM, West TE, Abd-Elsayed A (2022) Dorsal Root Ganglion Stimulation for Lower Extremity Neuropathic Pain Syndromes: An Evidence-Based Literature Review. Adv Ther 39(10):4440–4473. https://doi.org/10.1007/s12325-022-02244-9

    Article  PubMed  PubMed Central  Google Scholar 

  166. Ong Sio LC, Hom B, Garg S, Abd-Elsayed A (2023) Mechanism of Action of Peripheral Nerve Stimulation for Chronic Pain: A Narrative Review. Int J Mol Sci 24 (5). https://doi.org/10.3390/ijms24054540

  167. Mekhail NA, Estemalik E, Azer G, Davis K, Tepper SJ (2017) Safety and Efficacy of Occipital Nerves Stimulation for the Treatment of Chronic Migraines: Randomized, Double-blind. Controlled Single-center Experience Pain Pract 17(5):669–677. https://doi.org/10.1111/papr.12504

    Article  PubMed  Google Scholar 

  168. Dodick DW, Silberstein SD, Reed KL, Deer TR, Slavin KV, Huh B, Sharan AD, Narouze S, Mogilner AY, Trentman TL, Ordia J, Vaisman J, Goldstein J, Mekhail N (2015) Safety and efficacy of peripheral nerve stimulation of the occipital nerves for the management of chronic migraine: long-term results from a randomized, multicenter, double-blinded, controlled study. Cephalalgia 35(4):344–358. https://doi.org/10.1177/0333102414543331

    Article  PubMed  Google Scholar 

  169. Silberstein SD, Dodick DW, Saper J, Huh B, Slavin KV, Sharan A, Reed K, Narouze S, Mogilner A, Goldstein J, Trentman T, Vaisman J, Ordia J, Weber P, Deer T, Levy R, Diaz RL, Washburn SN, Mekhail N (2012) Safety and efficacy of peripheral nerve stimulation of the occipital nerves for the management of chronic migraine: results from a randomized, multicenter, double-blinded, controlled study. Cephalalgia 32(16):1165–1179. https://doi.org/10.1177/0333102412462642

    Article  PubMed  Google Scholar 

  170. Saper JR, Dodick DW, Silberstein SD, McCarville S, Sun M, Goadsby PJ (2011) Occipital nerve stimulation for the treatment of intractable chronic migraine headache: ONSTIM feasibility study. Cephalalgia 31(3):271–285. https://doi.org/10.1177/0333102410381142

    Article  PubMed  PubMed Central  Google Scholar 

  171. Warner NS, Schaefer KK, Eldrige JS, Lamer TJ, Pingree MJ, Bendel MA, Warner MA, Rho RH, Mauck WD (2021) Peripheral Nerve Stimulation and Clinical Outcomes: A Retrospective Case Series. Pain Pract 21(4):411–418. https://doi.org/10.1111/papr.12968

    Article  PubMed  Google Scholar 

  172. Colini Baldeschi G, Dario A, De Carolis G, Luxardo N, Natale M, Nosella P, Papa A, Raggi M, Reverberi C (2017) Peripheral Nerve Stimulation in the Treatment of Chronic Pain Syndromes From Nerve Injury: A Multicenter Observational Study. Neuromodulation 20(4):369–374. https://doi.org/10.1111/ner.12539

    Article  PubMed  Google Scholar 

  173. Ellis JA, Mejia Munne JC, Winfree CJ (2015) Trigeminal branch stimulation for the treatment of intractable craniofacial pain. J Neurosurg 123(1):283–288. https://doi.org/10.3171/2014.12.JNS14645

    Article  PubMed  Google Scholar 

  174. Schoenen J, Jensen RH, Lanteri-Minet M, Lainez MJ, Gaul C, Goodman AM, Caparso A, May A (2013) Stimulation of the sphenopalatine ganglion (SPG) for cluster headache treatment. Pathway CH-1: a randomized, sham-controlled study. Cephalalgia. 33(10):816–830. https://doi.org/10.1177/0333102412473667

    Article  PubMed  PubMed Central  Google Scholar 

  175. Strand N, D’Souza RS, Hagedorn JM, Pritzlaff S, Sayed D, Azeem N, Abd-Elsayed A, Escobar A, Huntoon MA, Lam CM, Deer TR (2022) Evidence-Based Clinical Guidelines from the American Society of Pain and Neuroscience for the Use of Implantable Peripheral Nerve Stimulation in the Treatment of Chronic Pain. J Pain Res 15:2483–2504. https://doi.org/10.2147/JPR.S362204

    Article  PubMed  PubMed Central  Google Scholar 

  176. Vatansever D, Tekin I, Tuglu I, Erbuyun K, Ok G (2008) A comparison of the neuroablative effects of conventional and pulsed radiofrequency techniques. Clin J Pain 24(8):717–724. https://doi.org/10.1097/AJP.0b013e318173c27a

    Article  PubMed  Google Scholar 

  177. West M, Wu H (2010) Pulsed radiofrequency ablation for residual and phantom limb pain: a case series. Pain Pract 10(5):485–491. https://doi.org/10.1111/j.1533-2500.2009.00353.x

    Article  PubMed  Google Scholar 

  178. Park D, Chang MC (2022) The mechanism of action of pulsed radiofrequency in reducing pain: a narrative review. J Yeungnam Med Sci 39(3):200–205. https://doi.org/10.12701/jyms.2022.00101

    Article  PubMed  PubMed Central  Google Scholar 

  179. Huang RY, Liao CC, Tsai SY, Yen CT, Lin CW, Chen TC, Lin WT, Chang CH, Wen YR (2017) Rapid and Delayed Effects of Pulsed Radiofrequency on Neuropathic Pain: Electrophysiological, Molecular, and Behavioral Evidence Supporting Long-Term Depression. Pain Physician 20(2):E269–E283

    PubMed  Google Scholar 

  180. Sluijter ME, van Kleef M (2007) Pulsed radiofrequency. Pain Med 8(4):388–389. https://doi.org/10.1111/j.1526-4637.2007.00304.x. (author reply 390-381)

    Article  PubMed  Google Scholar 

  181. Lin CS, Lin YC, Lao HC, Chen CC (2019) Interventional Treatments for Postherpetic Neuralgia: A Systematic Review. Pain Physician 22(3):209–228

    Article  PubMed  Google Scholar 

  182. Ke M, Yinghui F, Yi J, Xeuhua H, Xiaoming L, Zhijun C, Chao H, Yingwei W (2013) Efficacy of pulsed radiofrequency in the treatment of thoracic postherpetic neuralgia from the angulus costae: a randomized, double-blinded, controlled trial. Pain Physician 16(1):15–25

    PubMed  Google Scholar 

  183. Pi ZB, Lin H, He GD, Cai Z, Xu XZ (2015) Randomized and controlled prospective trials of Ultrasound-guided spinal nerve posterior ramus pulsed radiofrequency treatment for lower back post-herpetic neuralgia. Clin Ter 166(5):e301-305. https://doi.org/10.7417/T.2015.1882

    Article  CAS  PubMed  Google Scholar 

  184. Wang D, Zhang K, Han S, Yu L (2017) PainVision(R) Apparatus for Assessment of Efficacy of Pulsed Radiofrequency Combined with Pharmacological Therapy in the Treatment of Postherpetic Neuralgia and Correlations with Measurements. Biomed Res Int 2017:5670219. https://doi.org/10.1155/2017/5670219

    Article  PubMed  PubMed Central  Google Scholar 

  185. Saxena AK, Lakshman K, Sharma T, Gupta N, Banerjee BD, Singal A (2016) Modulation of serum BDNF levels in postherpetic neuralgia following pulsed radiofrequency of intercostal nerve and pregabalin. Pain Manag 6(3):217–227. https://doi.org/10.2217/pmt.16.3

    Article  PubMed  Google Scholar 

  186. Park S, Park JH, Jang JN, Choi SI, Song Y, Kim YU (2024) Pulsed radiofrequency of lumbar dorsal root ganglion for lumbar radicular pain: a systematic review and meta-analysis. Pain Pract. https://doi.org/10.1111/papr.13351

    Article  PubMed  Google Scholar 

  187. Facchini G, Spinnato P, Guglielmi G, Albisinni U, Bazzocchi A (2017) A comprehensive review of pulsed radiofrequency in the treatment of pain associated with different spinal conditions. Br J Radiol 90(1073):20150406. https://doi.org/10.1259/bjr.20150406

    Article  PubMed  PubMed Central  Google Scholar 

  188. Deer T, Shah A, Slavin K, Vorenkamp KE, Shah S, Leong M, McRoberts WP (2023) Birds of a Feather Redux: Defining Ways to Stimulate the Peripheral Nervous System. J Pain Res 16:1219–1224. https://doi.org/10.2147/JPR.S409158

    Article  PubMed  PubMed Central  Google Scholar 

  189. Plaza-Manzano G, Gomez-Chiguano GF, Cleland JA, Arias-Buria JL, Fernandez-de-Las-Penas C, Navarro-Santana MJ (2020) Effectiveness of percutaneous electrical nerve stimulation for musculoskeletal pain: a systematic review and meta-analysis. Eur J Pain 24(6):1023–1044. https://doi.org/10.1002/ejp.1559

    Article  PubMed  Google Scholar 

  190. Kopsky DJ, Ettema FW, van der Leeden M, Dekker J, Stolwijk-Swuste JM (2014) Percutaneous nerve stimulation in chronic neuropathic pain patients due to spinal cord injury: a pilot study. Pain Pract 14(3):252–259. https://doi.org/10.1111/papr.12064

    Article  PubMed  Google Scholar 

  191. Ghoname EA, Craig WF, White PF, Ahmed HE, Hamza MA, Henderson BN, Gajraj NM, Huber PJ, Gatchel RJ (1999) Percutaneous electrical nerve stimulation for low back pain: a randomized crossover study. JAMA 281(9):818–823. https://doi.org/10.1001/jama.281.9.818

    Article  CAS  PubMed  Google Scholar 

  192. Raphael JH, Raheem TA, Southall JL, Bennett A, Ashford RL, Williams S (2011) Randomized double-blind sham-controlled crossover study of short-term effect of percutaneous electrical nerve stimulation in neuropathic pain. Pain Med 12(10):1515–1522. https://doi.org/10.1111/j.1526-4637.2011.01215.x

    Article  PubMed  Google Scholar 

  193. Rodriguez Lagos L, Arribas-Romano A, Fernandez-Carnero J, Gonzalez-Zamorano Y, Laguarta Val S (2023) Effects of Percutaneous and Transcutaneous Electrical Nerve Stimulation on Endogenous Pain Mechanisms in Patients with Musculoskeletal Pain: A Systematic Review and Meta-Analysis. Pain Med 24(4):397–414. https://doi.org/10.1093/pm/pnac140

    Article  PubMed  Google Scholar 

  194. Beltran-Alacreu H, Serrano-Munoz D, Martin-Caro Alvarez D, Fernandez-Perez JJ, Gomez-Soriano J, Avendano-Coy J (2022) Percutaneous Versus Transcutaneous Electrical Nerve Stimulation for the Treatment of Musculoskeletal Pain. A Systematic Review and Meta-Analysis. Pain Med 23(8):1387–1400. https://doi.org/10.1093/pm/pnac027

    Article  PubMed  Google Scholar 

  195. Lefaucheur JP, Antal A, Ayache SS, Benninger DH, Brunelin J, Cogiamanian F, Cotelli M, De Ridder D, Ferrucci R, Langguth B, Marangolo P, Mylius V, Nitsche MA, Padberg F, Palm U, Poulet E, Priori A, Rossi S, Schecklmann M, Vanneste S, Ziemann U, Garcia-Larrea L, Paulus W (2017) Evidence-based guidelines on the therapeutic use of transcranial direct current stimulation (tDCS). Clin Neurophysiol 128(1):56–92. https://doi.org/10.1016/j.clinph.2016.10.087

    Article  PubMed  Google Scholar 

  196. Nitsche MA, Cohen LG, Wassermann EM, Priori A, Lang N, Antal A, Paulus W, Hummel F, Boggio PS, Fregni F, Pascual-Leone A (2008) Transcranial direct current stimulation: State of the art 2008. Brain Stimul 1(3):206–223. https://doi.org/10.1016/j.brs.2008.06.004

    Article  PubMed  Google Scholar 

  197. DaSilva AF, Truong DQ, DosSantos MF, Toback RL, Datta A, Bikson M (2015) State-of-art neuroanatomical target analysis of high-definition and conventional tDCS montages used for migraine and pain control. Front Neuroanat 9:89. https://doi.org/10.3389/fnana.2015.00089

    Article  PubMed  PubMed Central  Google Scholar 

  198. Baptista AF, Fernandes A, Sa KN, Okano AH, Brunoni AR, Lara-Solares A, Jreige Iskandar A, Guerrero C, Amescua-Garcia C, Kraychete DC, Caparelli-Daquer E, Atencio E, Piedimonte F, Colimon F, Hazime FA, Garcia JBS, Hernandez-Castro JJ, Cantisani JAF, Karina do Monte-Silva K, Lemos Correia LC, Gallegos MS, Marcolin MA, Ricco MA, Cook MB, Bonilla P, Schestatsky P, Galhardoni R, Silva V, Delgado Barrera W, Caumo W, Bouhassira D, Chipchase LS, Lefaucheur JP, Teixeira MJ, de Andrade DC, (2019) Latin American and Caribbean consensus on noninvasive central nervous system neuromodulation for chronic pain management (LAC(2)-NIN-CP). Pain Rep 4(1):e692. https://doi.org/10.1097/PR9.0000000000000692

    Article  PubMed  PubMed Central  Google Scholar 

  199. Alwardat M, Pisani A, Etoom M, Carpenedo R, Chine E, Dauri M, Leonardis F, Natoli S (2020) Is transcranial direct current stimulation (tDCS) effective for chronic low back pain? A systematic review and meta-analysis. J Neural Transm (Vienna) 127(9):1257–1270. https://doi.org/10.1007/s00702-020-02223-w

    Article  PubMed  Google Scholar 

  200. Stilling JM, Monchi O, Amoozegar F, Debert CT (2019) Transcranial Magnetic and Direct Current Stimulation (TMS/tDCS) for the Treatment of Headache: A Systematic Review. Headache 59(3):339–357. https://doi.org/10.1111/head.13479

    Article  PubMed  Google Scholar 

  201. Hamid P, Malik BH, Hussain ML (2019) Noninvasive Transcranial Magnetic Stimulation (TMS) in Chronic Refractory Pain: A Systematic Review. Cureus 11(10):e6019. https://doi.org/10.7759/cureus.6019

    Article  PubMed  PubMed Central  Google Scholar 

  202. Barker AT, Jalinous R, Freeston IL (1985) Non-invasive magnetic stimulation of human motor cortex. Lancet 1(8437):1106–1107. https://doi.org/10.1016/s0140-6736(85)92413-4

    Article  CAS  PubMed  Google Scholar 

  203. Garcia-Larrea L (2023) Non-invasive cortical stimulation for drug-resistant pain. Curr Opin Support Palliat Care 17(3):142–149. https://doi.org/10.1097/SPC.0000000000000654

    Article  PubMed  Google Scholar 

  204. Lefaucheur JP, Andre-Obadia N, Antal A, Ayache SS, Baeken C, Benninger DH, Cantello RM, Cincotta M, de Carvalho M, De Ridder D, Devanne H, Di Lazzaro V, Filipovic SR, Hummel FC, Jaaskelainen SK, Kimiskidis VK, Koch G, Langguth B, Nyffeler T, Oliviero A, Padberg F, Poulet E, Rossi S, Rossini PM, Rothwell JC, Schonfeldt-Lecuona C, Siebner HR, Slotema CW, Stagg CJ, Valls-Sole J, Ziemann U, Paulus W, Garcia-Larrea L (2014) Evidence-based guidelines on the therapeutic use of repetitive transcranial magnetic stimulation (rTMS). Clin Neurophysiol 125(11):2150–2206. https://doi.org/10.1016/j.clinph.2014.05.021

    Article  PubMed  Google Scholar 

  205. Peinemann A, Reimer B, Loer C, Quartarone A, Munchau A, Conrad B, Siebner HR (2004) Long-lasting increase in corticospinal excitability after 1800 pulses of subthreshold 5 Hz repetitive TMS to the primary motor cortex. Clin Neurophysiol 115(7):1519–1526. https://doi.org/10.1016/j.clinph.2004.02.005

    Article  PubMed  Google Scholar 

  206. Bestmann S, Baudewig J, Siebner HR, Rothwell JC, Frahm J (2004) Functional MRI of the immediate impact of transcranial magnetic stimulation on cortical and subcortical motor circuits. Eur J Neurosci 19(7):1950–1962. https://doi.org/10.1111/j.1460-9568.2004.03277.x

    Article  PubMed  Google Scholar 

  207. Fierro B, De Tommaso M, Giglia F, Giglia G, Palermo A, Brighina F (2010) Repetitive transcranial magnetic stimulation (rTMS) of the dorsolateral prefrontal cortex (DLPFC) during capsaicin-induced pain: modulatory effects on motor cortex excitability. Exp Brain Res 203(1):31–38. https://doi.org/10.1007/s00221-010-2206-6

    Article  PubMed  Google Scholar 

  208. Yoo WK, You SH, Ko MH, Tae Kim S, Park CH, Park JW, Hoon Ohn S, Hallett M, Kim YH (2008) High frequency rTMS modulation of the sensorimotor networks: behavioral changes and fMRI correlates. Neuroimage 39(4):1886–1895. https://doi.org/10.1016/j.neuroimage.2007.10.035

    Article  PubMed  Google Scholar 

  209. Stephens E, Dhanasekara CS, Montalvan V, Zhang B, Bassett A, Hall R, Rodaniche A, Robohm-Leavitt C, Shen CL, Kahatuduwa CN (2024) Utility of Repetitive Transcranial Magnetic Stimulation for Chronic Daily Headache Prophylaxis: A Systematic Review and Meta-Analysis. Curr Pain Headache Rep. https://doi.org/10.1007/s11916-024-01210-0

    Article  PubMed  Google Scholar 

  210. Knorst GRS, Souza PR, Araujo A, Knorst SAF, Diniz DS, Filho H (2024) Transcranial magnetic stimulation in the treatment of phantom limb pain: a systematic review. Arq Neuropsiquiatr 82(1):1–10. https://doi.org/10.1055/s-0044-1779051

    Article  PubMed  PubMed Central  Google Scholar 

  211. Tiwari VK, Kumar A, Nanda S, Chaudhary S, Sharma R, Kumar U, Kumaran SS, Bhatia R (2024) Effect of neuronavigated repetitive Transcranial Magnetic Stimulation on pain, cognition and cortical excitability in fibromyalgia syndrome. Neurol Sci. https://doi.org/10.1007/s10072-024-07317-x

    Article  PubMed  Google Scholar 

  212. Ciampi de Andrade D, Garcia-Larrea L (2023) Beyond trial-and-error: Individualizing therapeutic transcranial neuromodulation for chronic pain. Eur J Pain 27(9):1065–1083. https://doi.org/10.1002/ejp.2164

    Article  PubMed  Google Scholar 

  213. Andre-Obadia N, Mertens P, Lelekov-Boissard T, Afif A, Magnin M, Garcia-Larrea L (2014) Is Life better after motor cortex stimulation for pain control? Results at long-term and their prediction by preoperative rTMS. Pain Physician 17(1):53–62

    Article  PubMed  Google Scholar 

  214. Andre-Obadia N, Peyron R, Mertens P, Mauguiere F, Laurent B, Garcia-Larrea L (2006) Transcranial magnetic stimulation for pain control Double-blind study of different frequencies against placebo, and correlation with motor cortex stimulation efficacy. Clin Neurophysiol 117(7):1536–1544. https://doi.org/10.1016/j.clinph.2006.03.025

    Article  PubMed  Google Scholar 

  215. Sluka KA, Walsh D (2003) Transcutaneous electrical nerve stimulation: basic science mechanisms and clinical effectiveness. J Pain 4(3):109–121. https://doi.org/10.1054/jpai.2003.434

    Article  PubMed  Google Scholar 

  216. Cheing GL, Hui-Chan CW (1999) Transcutaneous electrical nerve stimulation: nonparallel antinociceptive effects on chronic clinical pain and acute experimental pain. Arch Phys Med Rehabil 80(3):305–312. https://doi.org/10.1016/s0003-9993(99)90142-9

    Article  CAS  PubMed  Google Scholar 

  217. Deyo RA, Walsh NE, Martin DC, Schoenfeld LS, Ramamurthy S (1990) A controlled trial of transcutaneous electrical nerve stimulation (TENS) and exercise for chronic low back pain. N Engl J Med 322(23):1627–1634. https://doi.org/10.1056/NEJM199006073222303

    Article  CAS  PubMed  Google Scholar 

  218. Verville L, Hincapie CA, Southerst D, Yu H, Bussieres A, Gross DP, Pereira P, Mior S, Tricco AC, Cedraschi C, Brunton G, Nordin M, Connell G, Shearer HM, Wong JJ, Hofstetter L, Romanelli A, Guist B, To D, Stuber K, da Silva-Oolup S, Stupar M, Myrtos D, Lee JGB, DeSouza A, Munoz Laguna J, Murnaghan K, Cancelliere C (2023) Systematic Review to Inform a World Health Organization (WHO) Clinical Practice Guideline: Benefits and Harms of Transcutaneous Electrical Nerve Stimulation (TENS) for Chronic Primary Low Back Pain in Adults. J Occup Rehabil 33(4):651–660. https://doi.org/10.1007/s10926-023-10121-7

    Article  PubMed  PubMed Central  Google Scholar 

  219. Dailey DL, Rakel BA, Vance CGT, Liebano RE, Amrit AS, Bush HM, Lee KS, Lee JE, Sluka KA (2013) Transcutaneous electrical nerve stimulation reduces pain, fatigue and hyperalgesia while restoring central inhibition in primary fibromyalgia. Pain 154(11):2554–2562. https://doi.org/10.1016/j.pain.2013.07.043

    Article  PubMed  PubMed Central  Google Scholar 

  220. Gikaro JM, Bigambo FM, Minde VM, Swai EA (2023) Efficacy of electrophysical agents in fibromyalgia: A systematic review and network meta-analysis. Clin Rehabil 37(10):1295–1310. https://doi.org/10.1177/02692155231170450

    Article  PubMed  Google Scholar 

  221. Schoenen JE (2016) Migraine prevention with a supraorbital transcutaneous stimulator: A randomized controlled trial. Neurology 86(2):201–202. https://doi.org/10.1212/01.wnl.0000479686.32453.cc

    Article  PubMed  Google Scholar 

  222. Chou DE, Shnayderman Yugrakh M, Winegarner D, Rowe V, Kuruvilla D, Schoenen J (2019) Acute migraine therapy with external trigeminal neurostimulation (ACME): A randomized controlled trial. Cephalalgia 39(1):3–14. https://doi.org/10.1177/0333102418811573

    Article  PubMed  Google Scholar 

  223. di Biase L, Falato E, Di Lazzaro V (2019) Transcranial Focused Ultrasound (tFUS) and Transcranial Unfocused Ultrasound (tUS) Neuromodulation: From Theoretical Principles to Stimulation Practices. Front Neurol 10:549. https://doi.org/10.3389/fneur.2019.00549

    Article  PubMed  PubMed Central  Google Scholar 

  224. Zhang T, Guo B, Zuo Z, Long X, Hu S, Li S, Su X, Wang Y, Liu C (2023) Excitatory-inhibitory modulation of transcranial focus ultrasound stimulation on human motor cortex. CNS Neurosci Ther 29(12):3829–3841. https://doi.org/10.1111/cns.14303

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Mishra A, Yang PF, Manuel TJ, Newton AT, Phipps MA, Luo H, Sigona MK, Reed JL, Gore JC, Grissom WA, Caskey CF, Chen LM (2023) Disrupting nociceptive information processing flow through transcranial focused ultrasound neuromodulation of thalamic nuclei. Brain Stimul 16(5):1430–1444. https://doi.org/10.1016/j.brs.2023.09.013

    Article  PubMed  PubMed Central  Google Scholar 

  226. Nussel M, Zhao Y, Knorr C, Regensburger M, Stadlbauer A, Buchfelder M, Del Vecchio A, Kinfe T (2022) Deep Brain Stimulation, Stereotactic Radiosurgery and High-Intensity Focused Ultrasound Targeting the Limbic Pain Matrix: A Comprehensive Review. Pain Ther 11(2):459–476. https://doi.org/10.1007/s40122-022-00381-1

    Article  PubMed  PubMed Central  Google Scholar 

  227. Todd N, McDannold N, Borsook D (2020) Targeted manipulation of pain neural networks: The potential of focused ultrasound for treatment of chronic pain. Neurosci Biobehav Rev 115:238–250. https://doi.org/10.1016/j.neubiorev.2020.06.007

    Article  PubMed  PubMed Central  Google Scholar 

  228. Meyerson BA (2001) Neurosurgical approaches to pain treatment. Acta Anaesthesiol Scand 45(9):1108–1113. https://doi.org/10.1034/j.1399-6576.2001.450910.x

    Article  CAS  PubMed  Google Scholar 

  229. Jeanmonod D, Werner B, Morel A, Michels L, Zadicario E, Schiff G, Martin E (2012) Transcranial magnetic resonance imaging-guided focused ultrasound: noninvasive central lateral thalamotomy for chronic neuropathic pain. Neurosurg Focus 32(1):E1. https://doi.org/10.3171/2011.10.FOCUS11248

    Article  PubMed  Google Scholar 

  230. Martin E, Jeanmonod D, Morel A, Zadicario E, Werner B (2009) High-intensity focused ultrasound for noninvasive functional neurosurgery. Ann Neurol 66(6):858–861. https://doi.org/10.1002/ana.21801

    Article  PubMed  Google Scholar 

  231. Richards DE, Tyner CF, Shealy CN (1966) Focused ultrasonic spinal commissurotomy: experimental evaluation. J Neurosurg 24(4):701–707. https://doi.org/10.3171/jns.1966.24.4.0701

    Article  CAS  PubMed  Google Scholar 

  232. Bouwense SA, Olesen SS, Drewes AM, van Goor H, Wilder-Smith OH (2015) Pregabalin and placebo responders show different effects on central pain processing in chronic pancreatitis patients. J Pain Res 8:375–386. https://doi.org/10.2147/JPR.S84484

    Article  PubMed  PubMed Central  Google Scholar 

  233. Jeon SY, Choi JH, Kang SS, An YH, Shim HJ (2023) Personalized Neuromodulation: A Novel Strategy for Improving Tinnitus Treatment. J Clin Med 12 (22). https://doi.org/10.3390/jcm12226987

  234. Schoisswohl S, Langguth B, Hebel T, Vielsmeier V, Abdelnaim MA, Schecklmann M (2022) Personalization of Repetitive Transcranial Magnetic Stimulation for the Treatment of Chronic Subjective Tinnitus. Brain Sci 12 (2). https://doi.org/10.3390/brainsci12020203

  235. Schoisswohl S, Langguth B, Hebel T, Abdelnaim MA, Volberg G, Schecklmann M (2021) Heading for Personalized rTMS in Tinnitus: Reliability of Individualized Stimulation Protocols in Behavioral and Electrophysiological Responses. J Pers Med 11 (6). https://doi.org/10.3390/jpm11060536

  236. Schoisswohl S, Langguth B, Schecklmann M (2020) Short-Term Tinnitus Suppression With Electric-Field Guided rTMS for Individualizing rTMS Treatment: A Technical Feasibility Report. Front Neurol 11:86. https://doi.org/10.3389/fneur.2020.00086

    Article  PubMed  PubMed Central  Google Scholar 

  237. Kreuzer PM, Poeppl TB, Rupprecht R, Vielsmeier V, Lehner A, Langguth B, Schecklmann M (2017) Individualized Repetitive Transcranial Magnetic Stimulation Treatment in Chronic Tinnitus? Front Neurol 8:126. https://doi.org/10.3389/fneur.2017.00126

    Article  PubMed  PubMed Central  Google Scholar 

  238. Boutet A, Madhavan R, Elias GJB, Joel SE, Gramer R, Ranjan M, Paramanandam V, Xu D, Germann J, Loh A, Kalia SK, Hodaie M, Li B, Prasad S, Coblentz A, Munhoz RP, Ashe J, Kucharczyk W, Fasano A, Lozano AM (2021) Predicting optimal deep brain stimulation parameters for Parkinson’s disease using functional MRI and machine learning. Nat Commun 12(1):3043. https://doi.org/10.1038/s41467-021-23311-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  239. Gilron R, Little S, Perrone R, Wilt R, de Hemptinne C, Yaroshinsky MS, Racine CA, Wang SS, Ostrem JL, Larson PS, Wang DD, Galifianakis NB, Bledsoe IO, San Luciano M, Dawes HE, Worrell GA, Kremen V, Borton DA, Denison T, Starr PA (2021) Long-term wireless streaming of neural recordings for circuit discovery and adaptive stimulation in individuals with Parkinson’s disease. Nat Biotechnol 39(9):1078–1085. https://doi.org/10.1038/s41587-021-00897-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Di Maio R (2014) Neuronal mechanisms of epileptogenesis. Front Cell Neurosci 8:29. https://doi.org/10.3389/fncel.2014.00029

    Article  PubMed  PubMed Central  Google Scholar 

  241. van Rooij SJH, Arulpragasam AR, McDonald WM, Philip NS (2024) Correction: Accelerated TMS - moving quickly into the future of depression treatment. Neuropsychopharmacology 49(1):297. https://doi.org/10.1038/s41386-023-01714-0

    Article  PubMed  Google Scholar 

  242. d'Andrea G, Mancusi G, Santovito MC, Marrangone C, Martino F, Santorelli M, Miuli A, Di Carlo F, Signorelli MS, Clerici M, Pettorruso M, Martinotti G (2023) Investigating the Role of Maintenance TMS Protocols for Major Depression: Systematic Review and Future Perspectives for Personalized Interventions. J Pers Med 13 (4). https://doi.org/10.3390/jpm13040697

  243. Andre-Obadia N, Mertens P, Gueguen A, Peyron R, Garcia-Larrea L (2008) Pain relief by rTMS: differential effect of current flow but no specific action on pain subtypes. Neurology 71(11):833–840. https://doi.org/10.1212/01.wnl.0000325481.61471.f0

    Article  CAS  PubMed  Google Scholar 

  244. Bastuji H, Cadic-Melchior A, Ruelle-Le Glaunec L, Magnin M, Garcia-Larrea L (2024) Functional connectivity between medial pulvinar and cortical networks as a predictor of arousal to noxious stimuli during sleep. Eur J Neurosci 59(4):570–583. https://doi.org/10.1111/ejn.15958

    Article  CAS  PubMed  Google Scholar 

  245. Radulescu I, Dragoi AM, Trifu SC, Cristea MB (2021) Neuroplasticity and depression: Rewiring the brain’s networks through pharmacological therapy (Review). Exp Ther Med 22(4):1131. https://doi.org/10.3892/etm.2021.10565

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  246. Sampathkumar V, Miller-Hansen A, Sherman SM, Kasthuri N (2021) Integration of signals from different cortical areas in higher order thalamic neurons. Proc Natl Acad Sci U S A 118 (30). https://doi.org/10.1073/pnas.2104137118

  247. Cury RG, Teixeira MJ, Galhardoni R, Silva V, Iglesio R, Franca C, Arnaut D, Fonoff ET, Barbosa ER, Ciampi de Andrade D (2020) Connectivity Patterns of Subthalamic Stimulation Influence Pain Outcomes in Parkinson’s Disease. Front Neurol 11:9. https://doi.org/10.3389/fneur.2020.00009

    Article  PubMed  PubMed Central  Google Scholar 

  248. Vetterlein A, Monzel M, Reuter M (2023) Are catechol-O-methyltransferase gene polymorphisms genetic markers for pain sensitivity after all? - A review and meta-analysis. Neurosci Biobehav Rev 148:105112. https://doi.org/10.1016/j.neubiorev.2023.105112

    Article  CAS  PubMed  Google Scholar 

  249. Hong H, Kim RY, Song Y, Suh C, Lee H, Lyoo IK, Yoon S, Lim SM, Lee S (2023) Genetic profile for dopamine signaling predicts brain functional reactivity to repetitive transcranial magnetic stimulation. Eur Arch Psychiatry Clin Neurosci 273(1):99–111. https://doi.org/10.1007/s00406-022-01436-2

    Article  PubMed  Google Scholar 

  250. Ojala J, Vanhanen J, Harno H, Lioumis P, Vaalto S, Kaunisto MA, Putaala J, Kangasniemi M, Kirveskari E, Makela JP, Kalso E (2022) A Randomized, Sham-Controlled Trial of Repetitive Transcranial Magnetic Stimulation Targeting M1 and S2 in Central Poststroke Pain: A Pilot Trial. Neuromodulation 25(4):538–548. https://doi.org/10.1111/ner.13496

    Article  PubMed  Google Scholar 

  251. Lindholm P, Lamusuo S, Taiminen T, Pesonen U, Lahti A, Virtanen A, Forssell H, Hietala J, Hagelberg N, Pertovaara A, Parkkola R, Jaaskelainen S (2015) Right secondary somatosensory cortex-a promising novel target for the treatment of drug-resistant neuropathic orofacial pain with repetitive transcranial magnetic stimulation. Pain 156(7):1276–1283. https://doi.org/10.1097/j.pain.0000000000000175

    Article  CAS  PubMed  Google Scholar 

  252. Motzkin JC, Kanungo I, D’Esposito M, Shirvalkar P (2023) Network targets for therapeutic brain stimulation: towards personalized therapy for pain. Front Pain Res (Lausanne) 4:1156108. https://doi.org/10.3389/fpain.2023.1156108

    Article  PubMed  Google Scholar 

  253. Fishman MA, Antony A, Esposito M, Deer T, Levy R (2019) The Evolution of Neuromodulation in the Treatment of Chronic Pain: Forward-Looking Perspectives. Pain Med 20(Suppl 1):S58–S68. https://doi.org/10.1093/pm/pnz074

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

none.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

GG, ADT, FL and DLT substantial contributed to the conception and design of the work. All authors acquired data and information from the study literature, and they wrote the drafted manuscript. GG, ADT, FL and DLT wrote and revised the final manuscript, and they critically reviewed it for scientific and intellectual content. All authors agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work were appropriately investigated and resolved.

Corresponding author

Correspondence to Federico Longhini.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

Prof. Federico Longhini contributed to the development of a new helmet for mechanical ventilation, and he is designated as inventor (European Patent number 3320941) not related to the present manuscript. He also received speaking fees from Draeger, Intersurgical and Fisher & Paykel. Prof. Federico Longhini acts also as Associate Editor for the Journal of Anesthesia, Analgesia and Critical Care. The remaining authors have no relevant financial or non-financial interests to disclose.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guzzi, G., Della Torre, A., Bruni, A. et al. Anatomo-physiological basis and applied techniques of electrical neuromodulation in chronic pain. J Anesth Analg Crit Care 4, 29 (2024). https://doi.org/10.1186/s44158-024-00167-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s44158-024-00167-1

Keywords