Next Article in Journal
Current and Emerging Therapies for Atherosclerotic Cardiovascular Disease Risk Reduction in Hypertriglyceridemia
Previous Article in Journal
An Overview of Systematic Reviews and Meta-Analyses on the Effect of Medication Interventions Targeting Polypharmacy for Frail Older Adults
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Diagnosis and Management of Neuropathic Pain in Spine Diseases

1
Department of Neurology, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland
2
Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland
3
Department of Clinical Immunology, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland
4
Department of Didactics and Medical Simulation, Medical University of Lublin, Chodźki 4, 20-093 Lublin, Poland
*
Author to whom correspondence should be addressed.
Submission received: 23 December 2022 / Revised: 31 January 2023 / Accepted: 7 February 2023 / Published: 9 February 2023
(This article belongs to the Section Clinical Neurology)

Abstract

:
Neuropathic pain is generally defined as a non-physiological pain experience caused by damage to the nervous system. It can occur spontaneously, as a reaction to a given stimulus, or independently of its action, leading to unusual pain sensations usually referred to as firing, burning or throbbing. In the course of spine disorders, pain symptoms commonly occur. According to available epidemiological studies, a neuropathic component of pain is often present in patients with spinal diseases, with a frequency ranging from 36% to 55% of patients. Distinguishing between chronic nociceptive pain and neuropathic pain very often remains a challenge. Consequently, neuropathic pain is often underdiagnosed in patients with spinal diseases. In reference to current guidelines for the treatment of neuropathic pain, gabapentin, serotonin and norepinephrine reuptake inhibitors and tricyclic antidepressants constitute first-line therapeutic agents. However, long-term pharmacologic treatment often leads to developing tolerance and resistance to used medications. Therefore, in recent years, a plethora of therapeutic methods for neuropathic pain have been developed and investigated to improve clinical outcomes. In this review, we briefly summarized current knowledge about the pathophysiology and diagnosis of neuropathic pain. Moreover, we described the most effective treatment approaches for neuropathic pain and discussed their relevance in the treatment of spinal pain.

1. Introduction

Neuropathic pain is generally defined as a non-physiological pain experience caused by damage to the nervous system [1]. It can occur in response to a given stimulus, independently of that stimulus, or spontaneously. Pain sensations reported by patients are usually referred as firing, burning or throbbing [2]. The etiology may be primary, when the pathological process involves the nervous tissue, or secondary, when the surrounding tissues are involved [3]. In many cases, it is difficult to distinguish the neuropathic component of pain reported by the patient, so neuropathic pain often occurs unnoticed by a physician. However, it is estimated that it affects about 7–8% of the general population [4].
Neuropathic pain is a clinical description, not a diagnosis. The states characteristic for the phenomenon of neuropathic pain are hyperalgesia, which is an excessive reaction to a given pain stimulus, and allodynia, defined as a feeling of pain resulting from the action of a stimulus that should not normally cause it [5]. Both suggest neuropathic pain, but are not necessary for its diagnosis.
There are many divisions depending on the mechanisms that lead to pain. Thus, there is central pain, when the damage leading to its occurrence is to the brain or spinal cord, and peripheral pain, originating from peripheral nerves, plexus and roots [6]. Interestingly, when the spinal cord is injured, up to half of the patients may experience neuropathic pain, while peripheral nerve injury is associated with a higher prevalence (approximately 73%) [7,8]. Another division distinguishes the various natures of the cause of the phenomenon, for example, mechanical, inflammatory, vascular or metabolic [9,10]. The same condition can cause severe pain in one patient without causing any in others, but the reasons for this differentiation remain unknown [2].
In the course of spine disorders, pain symptoms commonly occur [11,12]. According to available epidemiological studies, a neuropathic component of pain is often present in patients with spinal diseases, with a frequency ranging from 36% to 55% of patients [13,14]. Therefore, the use of therapeutic methods effective for neuropathic pain may be beneficial in the treatment of pain caused by spine disorders. However, molecular mechanisms participating in the pathomechanism of neuropathic pain are not fully understood, which may be the cause of the insufficient efficacy of the current treatment. Thus, to better investigate the pathomechanism of neuropathic pain and improve the effectiveness of therapy, an inverse translational approach has been recently introduced [15].
In this review, we briefly summarized current knowledge about the pathophysiology and diagnosis of neuropathic pain. Animal models of neuropathic pain, which are especially important today in the era of inverse translational approaches in neuropathic pain research, have also been presented. Moreover, we described the most effective treatment approaches for neuropathic pain and discussed their relevance in the treatment of spinal pain.

2. Pathophysiology of Neuropathic Pain

The mechanisms leading to the occurrence of pain are complex. Damage to the somatosensory system essentially leads to a loss of sensation, which is a so-called negative sensor sign [1]. However, some of the nerves may remain undamaged or partially damaged and are responsible for the conduction of not necessarily correct sensory feelings. Therefore, multiple processes within the lesion can simultaneously cause positive signs, of which ongoing pain is the most prominent one [1]. These phenomena may be caused either by increased neuronal transmission or by the weakening of the inhibitory processes, and also as a result of both of these processes. There is a frequent coexistence of both negative and positive sensory phenomena [16]. Understanding the pathologies responsible for the development of neuropathic pain is crucial in selecting the appropriate therapy.
The changes leading to the development of neuropathic pain include all levels of sensor transmission, the peripheral structures and their surroundings, as well as the central nervous system, and the processes on individual levels are sometimes slightly different. At the peripheral level, damage may be the cause of increased sensitization; secondarily, it may lead to ectopic impulses generation due to both degeneration in the affected area and subsequent regeneration [17]. These impulses, conducted via the normal nociceptive pathways, result in projected pain, which is a feeling of pain in the part of the body where the damage has occurred [18]. In addition, central sensitization and reorganization of the sensory fields can take place in the central nervous system, often as a result of microglia activity and excessive neurotransmission [17]. These changes, also sometimes manifested as projected pain, can secondarily cause further pathological processes in the more rostral structures of the nervous system [19]. Therefore, peripheral lesions leading to neuropathic pain lead to both peripheral and central changes, while lesions in the central nervous system are only characterized by the appearance of central changes.

2.1. Peripheral Mechanisms

Ectopic potentials seem to be the main mechanism responsible for the induction of peripheral neuropathic pain [20]. The spontaneous generation of potentials may be caused by a change in the expression of sodium channels in injured nerves [21]. The intact nerves necessary for conducting such signals are reportedly often subject to pathological sensitization within damaged tissue, where the expression of various growth factors that act on intact nerves is enhanced [22]. Ectopic generated potentials are conducted along the normal nerve pathways to the brain, which interprets them as pain from the territory of the body normally innervated by a given nerve, which underlies the phenomenon of projected pain [22].
Such stimulations conducted through the normal nociceptive pathways may induce use-dependent synaptic transmission [23]. This physiological phenomenon may be the first cause of central sensitization secondary to peripheral changes. The remaining reasons result from the above-mentioned over-expression of growth factors leading to changes in expression in the dorsal root ganglion and the axonal transport of chemokines that stimulate the spinal microglia [24]. Ultimately, central sensitization leads to increased activity of neurons receptive to normal or subliminal stimuli [23].
Therefore, in peripheral damage, pain is the result of both signals from the damaged tissue and conducted along the normal conduction pathways, and secondary changes in these pathways, also at the central level.

2.2. Central Mechanisms

The aforementioned central sensitization may be secondary to peripheral changes or result directly from pathology in the central nervous system. Synapses in the afferent pain pathways are subject to use-dependent plasticity [23]. These processes are similar to long-term potentiation and long-term depression in the hippocampus [25]. In contrast to the hippocampus, in the spinal cord, however, long-term potentiation-like phenomena can be induced by relatively low-frequency inputs, such as stimuli normally flowing into nociceptive neurons [26]. Many transmitters are involved in the formation of central sensitization and the loss or impairment of their functions may enhance central sensitization. Of importance are N-methyl-D-aspartate (NMDA) receptors for glutamate and neurokinin 1 (NK1) receptors for substance P, which are secreted presynaptically in nociceptive neurons [27]. Glutamate receptors of the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) type are highly regulated in the spinal cord plasticity processes [28]. Pain transmission is also regulated by a network of interneurons, some of which have inhibitory action through glycine or gamma-aminobutyric acid (GABA) [29].
Similar ectopic impulses can occur in the central nervous system, as in the case of peripheral pathologies. For example, bursting activity is observed in the somatosensory part of the thalamus and some animal models even suggest that this activity may be generated in the thalamus itself [30]. This is due to electrophysiological changes in the central nervous system, such as the upregulation of voltage-gated sodium channels [30]. Additionally, the decreased inhibition on the part of the GABAergic fibers may contribute to these disorders [31].
The perception of pain is physiologically regulated by descending fibers, both inhibitory and excitatory [19]. The role of rostroventral medulla is important here. Disorders of these mechanisms are also considered to be one of the central mechanisms responsible for the phenomenon of neuropathic pain, and although the reduced inhibition may play an important role in the phenomenon of hyperalgesia, it is suggested that the role of increased descending facilitation is much more important [32,33,34].
Another aspect observed in the case of neuropathic pain is the reorganization of the receptive field; however, it is not certain whether these mechanisms are one of the causes or a consequence of the existing pathology [35].

3. Animal Models of Neuropathic Pain

Neuropathic pain is a set of phenomena responsible for the occurrence of pain and other sensory pathologies that are inadequate to the stimulus that causes them, or even appear spontaneously regardless of this stimulus [36]. It includes such phenomena as hyperalgesia and allodynia [5]. These phenomena result, as already described, from peripheral and central causes, the mutual correlations of which depend on the location and nature of the primary pathology. Research on the nature of neuropathic pain is carried out in animal models that are highly differentiated in terms of the location of the damage on different levels of the afferent pathways from the peripheral nerve, through the spinal nerve to the spinal cord, and the type of damage, including traumatic, metabolic or toxic changes. The reconstruction of various disease states in animals, in which the phenomenon of neuropathic pain occurs, helps to understand the nature of the phenomenon, understand the role of particular pathologies that comprise the clinical picture, and search for therapeutic solutions that allow for effective treatment. It should be emphasized, however, that a large proportion of animal models only reflect single aspects, while clinically occurring neuropathic pain is a much more complex and diverse phenomenon, and a patient suffering from it may actually represent several phenomena simultaneously, composing a uniform picture of neuropathic pain. In all animal models described below, rats are the species of choice.

3.1. Chronic Constriction Injury

An important aspect of neuropathic pain is incomplete damage to the nerve structures. Although some of the fibers are completely damaged, the remaining partially damaged or even intact ones may conduct the impulses responsible for the occurrence of the discussed phenomena [22]. In the chronic constriction injury (CCI) model, this is achieved by placing loose sutures on the sciatic nerve [37]. As a result, some of the fibers degenerate, but the remaining, mostly afferent, fibers remain functional. Moreover, the use of chromic catgut provides additionally inflammatory components [38]. A few days after such surgery, symptoms typical of neuropathic pain can be observed in rats.
The phenomena of mechanical, chemical and thermal hyperalgesia and thermal allodynia are monitored and measured using special devices. Moreover, spontaneous pain sensations are demonstrated. Hyperalgesia lasts about several months and is often followed by hypoalgesia [39].
The undoubted advantage of this method is the wide spectrum of phenomena typical for neuropathic pain, which it causes, reflecting the potential clinical condition as effectively as possible. This makes the model a good field for testing and developing therapies. In this way, the effects of tricyclic antidepressants, such as amitriptyline, which reduces thermal and mechanical hyperalgesia, or clomipramine and desipramine, which act on mechanical hyperalgesia, have been proven [40,41]. Also in humans, antidepressants have proved to be effective as components of the treatment of diabetic or even post-herpetic neuropathy [42]. The action of antiepileptic drugs such as gabapentin has been similarly proven [43]. In a recent study on the CCI rat model, the anti-inflammatory and analgesic effects were demonstrated by quercetin, a plant flavonol [44]. Moreover, it has been observed that quercetin acts through the inhibition of the MAPK signaling pathway and stimulation of the AMPK pathway. However, the CCI model of neuropathic pain presents some imperfections, such as uncontrollable tension of the sutures (it may influence the amount of injured nerve fibers), demonstration of hyperalgesia (it is not presented by clinical neuropathic pain), and less intensified allodynia compared with other NP models [45].

3.2. Segmental Spinal Nerve Ligation

The difference between segmental nerve ligation (SNL) and CCI is that in SNL, not only one nerve is damaged, but the ligation takes place on a given segment of the spinal cord (L5 or L5 and L6 are injured, whereas the L4 spinal nerve remains intact) [45]. As a result, the sciatic nerve has both damaged (L5 and L6) and undamaged (L4) nerve fibers. Moreover, this NP model requires extensive surgical procedure exposing spinous processes at the L4-S2 levels [45]. Long-lasting ongoing pain, heat hyperalgesia, cold allodynia and mechanical allodynia have been observed after SNL [46].
SNL is widely used in preclinical research on the treatment of neuropathic pain. Drugs such as carbamazepine, gabapentin, felbamate and pregabalin have been studied [47]. Recently, adipose tissue-derived stem cells demonstrated effective action against the cold allodynia in a rat SNL model [48]. Similarly, the method was used for the purpose of a deeper understanding of the pathophysiology of the phenomenon: for example, the redistribution of sodium channels in undamaged axons [49]. Moreover, the SNL model showed that in central sensitization mechanisms, the CXCL12/CXCR4 signaling axis may be involved, which constitutes the potential aim of neuropathic pain treatment [50]. Nevertheless, muscle damage during the surgery (which may influence the pathologic mechanism) and a demanding surgical approach with avoiding any injury of the L4 nerve represent the main disadvantages of the SNL model [45].

3.3. Partial Sciatic Nerve Ligation

The partial nerve ligation (PNL) model is a modified version of the CCI where the dorsal part of the rats' nerve thickness (one third to one half) is ligated [51]. The model results in mechanical hyperalgesia and allodynia, and signs of spontaneous pain. These symptoms occur within hours after ligation and remain for about 7 months [45]. The PNL model imitates contusion rather than compression of the nerve [45].
The effectiveness of gabapentin against central sensitization and the prevention of thermal hyperalgesia by amitriptyline were proven in studies using PNL animal models [52,53]. Furthermore, icariin—a natural flavonoid obtained from the Epimedium species—investigated on the PNL model demonstrated the alleviation of neuropathic pain through anti-apoptotic and anti-inflammatory features [54]. Moreover, easy surgical procedure and high reproducibility are the main benefits of the PNL model [45].

3.4. Spared Nerve Injury

In the spared nerve injury (SNI) model, the sciatic nerve is exposed and the tibial nerve, as well as the common peroneal nerve, are sectioned [55]. The sural nerve remains intact [56]. In this model, mechanical allodynia and hyperalgesia, and cold allodynia as well as heat hyperalgesia, were observed 2–3 days after damage [55,57]. The mentioned symptoms disappear after at least 15 months in the case of rats and 30 days in the case of mice [45].
Based on this model, the relieving effect of gabapentin, amitriptyline and opioids has been proven [58,59,60]. The main advantages of the SNI model include high reproducibility, an easy surgical procedure, a short duration of pain symptoms, and a lack of influence on animal’s daily activity [61]. On the other hand, compared with other models, a decreased level of local inflammation is observed in the SNI model [56]. A recent study by He et al. developed a modified SNI model through injuring the sural and common peroneal nerves and keeping intact the tibial nerve [62]. This model, defined by authors as SNIt, exhibits less motor dysfunction compared with the traditional SNI model.

3.5. Diabetic Neuropathy

Several models of diabetic neuropathy have been developed because neuropathic pain is a fairly common component of it [63]. These include chemically, genetically and diet-induced diabetes. Diabetes induced by injecting the rats with pancreatic B-cell toxic compound streptozotocin or alloxan, as well as that induced by a high-fat diet, corresponds to a disease with a normal genetic background [64,65,66]. Thermal and mechanical hyperalgesia, and mechanical and chemical allodynia in the chemical model, as well as mechanical allodynia and thermal hyperalgesia in the case of diet-induced diabetes, have been observed here. In genetically determined diabetes, thermal hyperalgesia and mechanical allodynia have been observed [67].
Diabetic models have been used to study the effects of a number of analgesics, including pregabalin, diclofenac, TCAs and morphine, the latter two being effective [68,69,70,71].

3.6. Spinal Cord Injury and Neuropathic Pain

Spinal cord injury is often manifested by the coexistence of neuropathic pain [72]. Due to the multitude of mechanisms leading to it, there are many models that reflect the clinical condition of the patient, including spinal cord hemisection, spinal cord ischemia and spinal contusion [73,74,75].
Spinal cord hemisection corresponds to Brown-Sequard syndrome, and thermal and mechanical allodynia have been observed [73].
Ischemia of a specific segment of the spinal cord is achieved through the application of photo-reactives and the use of a laser. The reaction in the rats' vessels leads to occlusion followed by ischemia of the desired segment. Mechanical allodynia was among others observed in the research [74]. This model was used to determine the clinical efficacy of drugs such as morphine, tocainide, baclofen, pentobarbital and carbamazepine [76,77,78,79,80].
The spinal contusion model is achieved by dropping weights onto a fixed open spinal cord, which results in specific degrees of damage depending on the height from which the weights are dropped. The phenomena observed in this model are mechanical and thermal allodynia, and the model was used to deepen the knowledge of the pathophysiology of neuropathic pain with an indication of central pain [75]. Moreover, gabapentin has been proven to act against allodynia [81].

3.7. Visceral Pain Models

Models involving visceral pain include interstitial cystitis, endometriosis and prostatitis, the former of which is the most common in research [82,83,84]. Pain is often caused by the intraperitoneal administration of cyclophosphamide, which accumulates and undergoes metabolism, leading to inflammation and pain within 15–30 min of administration [85]. Another way to induce interstitial cystitis is by administering a pseudorabies virus, which is injected into the abductor caudalis dorsalis muscle [86]. Mast cell inflammation in this model suggests a strong association between neurogenic pain and inflammation.

4. Diagnosis and Frequency of Neuropathic Pain in Spine Diseases

Chronic pain is the primary symptom of patients suffering from spine diseases. Pain can vary in nature and severity depending on the mechanisms by which it is triggered. Although the nociceptive mechanism is the basic one, the neuropathic component sometimes seems to be equally important and may significantly affect the treatment and outcome of patients [87]. It should be noted that distinguishing between chronic inflammatory or nociceptive pain and neuropathic pain very often remains a challenge [88]. This is due to many factors, including the broad subjectivization of patients’ pain sensations, various dynamics of pain progression, the lack of an obvious clinical test to assist the diagnosis, as well as the imprecise definition of neuropathic pain [87].
A detailed anamnesis and medical examination increase the chance of a correct diagnosis. While nociceptive pain is usually sharp and rushing, neuropathic pain is stinging, tingling, numb, stabbing, and is accompanied by decreased sensitivity in the affected area [89,90]. There are no specific patterns of its course or triggers. It can appear spontaneously or be induced; sometimes it lasts constantly; other times it can be paroxysmal [90]. The occurrence of neuropathic pain may be indicated by the presence or history of a disease that could cause nerve damage. Moreover, the nerve damage confirmed by neurophysiological, neuroimaging, or during the surgery, in correlation with the overall assessment of the patient, allows for an almost certain diagnosis. The absence of a sensory disturbance and a good response to typically used analgesics make the diagnosis of neuropathic pain unlikely [90]. Various scales and questionnaires have been developed to help assess pain in patients and make the diagnosis of neuropathic pain, including the Leeds Assessment of Neuropathic Symptoms and Signs score (LANSS), the Northwick Park Neck Pain Questionnaire (NPQ), DN4 (Douleur Neuropathique 4 Questions) questionnaire, and painDETECT questionnaire [91,92].
Due to the aspects described above, neuropathic pain is often underdiagnosed in patients with spinal diseases. In the general population, the incidence of neuropathic pain is estimated to be from 0.82% to approximately 3%, while the overall incidence among chronic pain patients is estimated at 6.9% [93]. There are many studies assessing the incidence of neuropathic pain in patients with spine disease. For instance, Kim et al. described a group of 1109 patients who qualified for lumbar spine surgery, of whom 404 were diagnosed with neuropathic pain using the LANSS scale, which accounted for 36.4% of all assessed patients [94]. A very similar result on a smaller group of patients with lumbar spine stenosis was reported by Park et al. diagnosing neuropathic pain in 31 out of 86 patients (36%). However, this study divided patients complaining of radicular pain and neurogenic claudication, and in the former, the neuropathic component was much more frequent, occurring in 24 of them, which constituted 63.4%, while the remaining 7 with neurogenic claudication accounted for only 15.6%. It should be noted that the vast majority (78, which constituted 90.7%) of patients presented both root pain and claudication pain, and the above-mentioned division was made on the basis of the dominant component [95]. El Sissi et al. described the incidence of neuropathic pain in 55% of 1134 patients with chronic back pain [14]. Yamashita et al. reported a 53.5% frequency in patients with diseases of the spine [13]. Hassan et al. presented the prevalence of NP in 41% of the examined patients with back pain [96]. Attal et al. described the prevalence of neuropathic pain in patients, implying a higher occurrence correlated with lumbar spine stenosis then other causes [97,98] (Table 1).
The studies presented above prove how common the phenomenon of neuropathic pain is, which shows its clinical significance. As mentioned also, neuropathic pain is not usually responsive to standard NSAID analgesia. The response to drugs affecting opiate receptors is also sometimes limited. Antidepressants seem to be much more effective in the treatment of spinal diseases with a neuropathic component, including, for example, selective serotonin norepinephrine reuptake inhibitors (SSNRIs). Moreover, calcium channel α2-δ ligands and topical lidocaine might be useful [99,100,101,102,103]. This is one of the reasons why the diagnosis of neuropathic pain in diseases of the spine may play a key role in the selection of an effective therapy and contribute to a better outcome, especially in the treatment of patients with chronic pain not relieved by conventional therapy.

5. Treatment Strategies for Neuropathic Pain

In recent decades, a plethora of therapeutic methods for neuropathic pain have been developed and investigated. In the following sections, we describe currently recommended and emerging methods of treatment for neuropathic pain, and discuss their usefulness for the neuropathic component of chronic low back pain.

5.1. Pharmacotherapy

According to the recently updated (2020) French guidelines for the treatment of neuropathic pain, gabapentin, serotonin and norepinephrine reuptake inhibitors (SNRIs), and tricyclic antidepressants (TCAs), constitute first-line therapeutic agents [104]. On the other hand, pregabalin, tramadol, botulinum toxic A, capsaicin patches, combination therapies and psychotherapy constitutes are recommended as second-line therapy. Among third-line treatments, pharmacological agents such as strong opioids have also been considered. Moreover, gabapentinoids, SNRIs and TCAs were also proposed by NeuPSIG as first-line therapeutic agents [105]. These recommendations show the continuing major significance of pharmacotherapy in the management of neuropathic pain (Table 2).

5.1.1. Gabapentinoids

Pregabalin and gabapentin constitute the most common gabapentinoids prescribed in order to alleviate neuropathic pain in adults [139]. The gabapentinoids mechanism of action relies on binding to the α2-δ subunit of presynaptic voltage-gated Ca2+ channels in the dorsal horn of the spinal cord [106]. This leads to a decrease in the release of neurotransmitters, such as norepinephrine, glutamate and substance P. Consequently, that molecular mechanism is responsible for their analgesic effects [139]. Both gabapentin and pregabalin have demonstrated successful outcomes in the treatment of pain caused by postherpetic neuralgia, spinal cord injury, phantom limb syndrome and diabetes-induced neuropathy [108]. Most common complications observed in patients treated with gabapentionoids include peripheral edema, somnolence, dizziness, xerostomia and obesity [107].
Numerous scientific associations have recommended gabapentinoids as first-line drugs for neuropathic pain [104,105,140]. Moreover, the use of gabapentin and pregabalin for this purpose has been approved by the Food and Drug Administration (FDA) [108]. However, in a recent update of French guidelines, pregabalin was downgraded to a second-line treatment due to the risk of misuse and addiction, and a higher risk of respiratory depression in combination with opioids reported by the latest studies [129,141]. Moreover, a recent randomized controlled trial demonstrated that pregabalin was less safe and less effective in relieving sciatica compared with gabapentin [142].
Although pregabalin and gabapentin have demonstrated efficacy in the treatment of many types of neuropathic pain, their effectiveness in the treatment of neuropathic pain associated with spine diseases is not evident. Gabapentin has demonstrated significant improvements in sensory deficits, relieving pain and neurogenic claudication associated with lumbar spinal stenosis [143,144]. However, a recent meta-analysis did not recommended anticonvulsants as a first line treatment for lumbar spinal stenosis causing neurogenic claudication [145]. Regarding the use of gabapentinoids for sciatica, clinical trials evaluating their effectiveness in sciatica have showed unsatisfactory results [146,147,148]. A recent meta-analysis indicated that pregabalin and gabapentin did not provide improvement in sciatica compared with placebo and suggested their clinical use should not be supported for this purpose [149]. However, sciatica is characterized by a combination of nociceptive and neuropathic pain, where nociceptive pain constitutes the main component [150]. Therefore, gabapentionids, which cannot alleviate the nociceptive component of pain, may be ineffective for the management of sciatica.
A novel selective α2-δ ligand, mirogabalin, has shown safety and effectiveness in the management of postherpetic neuralgia and diabetic neuropathic pain [109]. Recently, mirogabalin has been approved for the treatment of peripheral neuropathic pain in Japan [151]. A retrospective study by Kim et al. evaluated mirogabalin in relieving neuropathic pain in 60 patients with lumbar spine disease [152]. Mild adverse effects in 17 patients and a significant improvement in leg pain symptoms were observed. Moreover, mirogabalin also alleviated low back pain and sleep disturbance. However, this study has some major limitations, such as no control group, a small study population and a retrospective design. In a multicenter randomized open-label study entitled MiroTAS, the addition of mirogabalin to the NSAIDs treatment of peripheral neuropathic pain related to lumbar spinal stenosis improved patients’ quality of life and decreased pain intensity compared with NSAIDs alone [153]. Although somnolence and dizziness were common adverse effects in the study group (30% and 25.5% respectively), the addition of mirogabalin to NSAIDs was considered a generally safe combination. Moreover, in another study, a decrease in somnolence and dizziness frequency was observed in patients who switched from pregabalin to mirogabalin (from 12.2% to 7.3% and from 14.6% to 4.9% respectively) [110]. The above findings suggest that mirogabalin may be a safer and similarly effective therapeutic approach for peripheral neuropathic pain in patients with spine diseases compared with other gabapentinoids, although further studies are necessary.

5.1.2. Antidepressants

Tricyclic antidepressants (TCAs) such as amitriptyline have been used as a treatment for neuropathic pain for many years and have been recommended as first-line drugs [140,141,154]. Their effectiveness has been proven in the treatment of postherpetic neuralgia, central postpartum pain, pain after spinal cord injury and nerve injury [105,108]. Recent studies have reported that TCAs also exhibit antitumoral effects and may be useful for the treatment of neuropathic pain associated with neoplastic disorders [155]. The exact mechanism underlying the analgesic properties of TCAs remains unclear. TCAs may inhibit neuropathic pain through increasing noradrenaline, which acts on α2-adrenergic receptors in the dorsal horn of the spinal cord. TCAs can also increase the level of noradrenaline in the locus coeruleus, which stimulates an impaired descending noradrenergic inhibitory system [111]. It is worth noting that the use of TCAs is contraindicated for patients suffering from cardiovascular disease, prostate hypertrophy and glaucoma [108,112]. Furthermore, cognitive impairment and walking disturbances may occur during therapy with TCAs [112]. Other often adverse effects reported in neuropathic pain studies in patients receiving TCAs include urinary retention, constipation, dry mouth and orthostatic hypotension [113]. However, the effects of long-term TCAs administration are not fully investigated in the available studies regarding the treatment of neuropathic pain, whereas severe withdrawal syndrome is a known long-term complication of therapy with TCAs [156].
According to a recent review, the use of TCAs in the treatment of low back pain can be successful [157]. However, the optimal dose of TCAs was not indicated in this study and was determined based on patient tolerance. Moreover, the low daily dose (10 mg) of TCAs such as amitriptyline or nortriptyline demonstrated the alleviation of leg and back pain in patients with lumbar spinal stenosis [158]. A meta-analysis by Ferreira et al. showed that TCAs were ineffective in reducing pain in patients with low back pain, but decreased pain intensity in patients with sciatica at a minimum of 2 weeks of follow-up [159]. However, the certainty of evidence obtained in this meta-analysis was low or even very low due to a limited number of patients, a high risk of bias, and imprecise estimates present in the included studies.
Duloxetine and venlafaxine are the most studied serotonin and norepinephrine reuptake inhibitors (SNRIs) for neuropathic pain treatment. These agents have been approved by the FDA for the treatment of fibromyalgia, diabetic neuropathy and back pain [114,115,116]. Due to the similarity of effects exerted by SNRIs to TCAs ones, their analgesic properties possibly result from increasing the level of noradrenaline in the dorsal horn of the spinal cord and locus coeruleus [111]. Common adverse reactions caused by SNRIs application include lack of appetite, constipation, dry mouth, anxiety, hyperhidrosis and nausea [117]. Moreover, SNRIs should be used with caution in patients with cardiovascular disease [160]. It is worth noting that the use of venlafaxine is associated with withdrawal syndrome [161].
In the treatment of chronic low back pain, the effectiveness of duloxetine is comparable to other oral pharmacological agents [162,163]. In a double-blind randomized controlled trial investigating the use of duloxetine in chronic low back pain with the neuropathic component, duloxetine significantly decreased pain symptoms by an average of 32% compared with placebo [164]. According to current studies, the optimal daily dose of 60 mg of duloxetine demonstrates the highest effectiveness in relieving pain, with the risk of complication reduced to a minimum [165]. Moreover, a recent retrospective study demonstrated the effectiveness of duloxetine in reducing postsurgical chronic neuropathic disorders in 19 of 24 patients after spine or spinal cord surgeries [166]. However, due to retrospective character and a limited number of patients in this clinical trial, the optimal use of duloxetine in patients with spine surgery-induced neuropathic pain should be investigated in further, well-designed randomized studies.

5.1.3. Topical Agents

Five-percent lidocaine patches have been indicated as a second-line agent by NeuPSIG recommendations and as a first-line agent for neuropathic pain according to recent French guidelines [105,140,141]. Lidocaine's mechanism of action relies on blocking voltage-gated Na+ channels, which decrease spontaneous nerve discharge [120]. Topical lidocaine is considered as the safest treatment alternative for neuropathic pain, and skin irritation constitutes its main adverse effect [119]. Numerous studies have demonstrated the efficacy of topical lidocaine in the treatment of postherpetic neuralgia, postsurgical chronic neuropathic pain and diabetic peripheral neuropathy [118]. For the treatment of pain caused by spine diseases, lidocaine can be administered both in the form of patches and paraspinal injections. Most of the studies published to date concerning lidocaine patches for chronic low back pain present little evidence due to their uncontrolled and open-label character [167]. A randomized controlled trial by Hashmi et al. demonstrated the non-superiority of a 5% lidocaine patch in the alleviation of chronic back pain compared with a placebo [168]. Therefore, the clinical efficacy of lidocaine patches in the treatment of low back pain is questionable. On the other hand, paraspinous lidocaine injections in combination with standard treatment demonstrated better outcomes in relieving low back pain compared with standard treatment alone, without an increased risk of adverse effects [169].
Topical capsaicin is generally recommended as a second-line drug, as neuropathic pain exerts its analgesic action through the modulation of transient receptor potential cation channel vanilloid subfamily member 1 (TRPV1), which is involved in the pain modulation mechanisms [105,140,141,170]. Similar to topical lidocaine, capsaicin is absorbed into the circulation at a minimum, and transient skin reactions dominate the adverse effects of this agent [121]. However, its potential long-term influence on nerve fibers is unknown [112]. The efficacy of 8% capsaicin patches was established in diabetic peripheral neuropathy, but beneficial outcomes were also reported in the treatment of postherpetic neuralgia and HIV neuropathies [122]. Although the number of clinical studies evaluating topical capsaicin in the treatment of neuropathic pain caused by spine diseases is limited, their results are promising. A prospective open-label clinical trial reported a significant alleviation of neuropathic pain and improvement in the quality of life of patients with lumbosacral neuropathic pain after the administration of an 8% capsaicin patch, compared with placebo [171]. Moreover, a significant decrease in neuropathic pain after the use of an 8% capsaicin patch has also been observed in patients with low back radiculopathies [172]. However, further randomized studies are necessary to verify the effectiveness of topical capsaicin for spinal neuropathic pain and to compare it with other treatment modalities.
Botulinum toxin A (BTX-A), a potent neurotoxin, is currently recommended as a second-line treatment for neuropathic pain [104]. Changes provoked by subcutaneously injected BTX-A include the modulation of both peripheral and central sensitizations responsible for the neuropathic pain pathomechanism [127]. The clinical effectiveness of BTX-A was demonstrated in several randomized controlled trials in the treatment of neuropathic pain forms, such as trigeminal neuralgia, postherpetic neuralgia, diabetic peripheral neuropathy and pain after spinal cord injury [123,124,125,126]. Moreover, BTX-A injections demonstrated better effects in the treatment of chronic low back pain compared with saline injections, a combination of lidocaine and corticosteroids, and traditional acupuncture [173]. In a recent prospective open-label study, BTX-A injection significantly improved pain scores measured in patients with resistant chronic low back pain with mild transient complications in a minority of patients [128]. The above findings should be confirmed by well-designed studies with a larger sample size.

5.1.4. Opioids

Opioids such as tramadol, tapentadol and oxycodone have also demonstrated potential effectiveness in relieving neuropathic pain with better results in peripheral than central neuropathic pain [112,129]. The high risk of misuse, addiction and morbidity comprise the main concerns of this class of drugs [131]. Tramadol, a µ-opioid agonist and 5-HT reuptake inhibitor, has been recommended as second-line treatment [108,112,141]. Somnolence and confusion may occur during treatment with tramadol, however, the risk of abuse is lower than other opioids [130]. Tapentadol, another µ-opioid agonist, has shown beneficial outcomes in the management of diabetic neuropathy and low back pain with a neuropathic component [174,175,176,177]. Other strong opioids such as oxycodone or morphine are recommended in the third-line treatment of neuropathic pain, but these agents are rarely utilized due to their high addiction potential [112]. A recent Bayesian network meta-analysis based on 2933 patients from published randomized controlled trials demonstrated that tapentadol, oxymorphone and fentanyl are the most effective opioids for pain symptoms in patients with chronic low back pain [178]. However, tramadol was not considered in this meta-analysis.

5.2. Neurostimulation Techniques

Long-term pharmacologic treatment often leads to developing tolerance and resistance to used medications. In the case of patients with neuropathic pain refractory to currently recommended pharmacologic treatment, CNS stimulation techniques may be a valuable therapeutic approach. These methods include spinal cord stimulation (SCS), dorsal root ganglion stimulation (DRGS), transcranial direct current stimulation (tDCS) and repetitive transcranial magnetic stimulation (rTMS). According to current French guidelines, SCS and rTMS are considered as a third-line treatment, whereas other neuromodulatory methods are not included [104] (Table 3).

5.2.1. Spinal Cord Stimulation

Among the investigated neuromodulatory techniques, spinal cord stimulation (SCS) constitutes a well-established method, which has demonstrated potential effectiveness in the treatment of numerous diseases such as peripheral diabetic neuropathy, failed back surgery syndrome, irritable bowel syndrome, painful radiculopathy, complex regional pain syndrome and postherpetic neuralgia [179,182,183,184,185,186]. Moreover, the cost-effectiveness of this method compared with conventional treatment modalities has been demonstrated [201].
In the SCS technique, multiple electrodes (usually between 4 and 16) are implanted percutaneously or through the laminotomy into the epidural space nearby the posterior columns of the spinal cord [202]. Thereafter, a low-voltage electrical current is administered through inserted electrodes at a frequency of about 50–60 Hz [108]. The electrical stimulation alleviates the symptoms of neuropathic pain due to the interruption of the nociceptive transmission to the brain through the inhibition of the C and Aδ-fibers [187]. Apart from neurophysiological mechanism, SCS also acts through an increase in GABA and serotonin levels and a subsequent decrease in the release of glutamate and aspartate, which also contributes to the analgesic effect exerted by SCS [188].
However, stimulation of the afferent sensory fibers with a large diameter such as Aδ-fibers may induce paresthesia. Therefore, numerous modifications of the SCS technique have been investigated with different stimulation parameters, which may prevent SCS-induced paresthesia [141,203]. One of them, a burst SCS, which relies on the administration of five intermittent electrical pulses at a frequency of around 500 Hz, effectively alleviated the pain caused by failed back surgery syndrome and painful diabetic neuropathy, and significantly decreased paresthesia compared with tonic SCS [204]. Based on the results of a systematic review, a I/II level of evidence was determined for the efficacy of SCS, and a II/III level of evidence for high-frequency stimulation in the treatment of lumbar FBSS [205]. Moreover, SCS significantly reduces chronic low back pain regardless of a history of previous spine surgery [202,206].
Due to the invasiveness of the epidural implantation of electrodes, numerous complications are associated with SCS, including pain at the generator site (12% of cases), infection (4.5% of cases), nerve injury and epidural hematoma [112,180,189]. Moreover, migration, breakage, disconnection or failure of the electrode may complicate the treatment by SCS [181].
Nevertheless, SCS represents a relatively well-tolerated, effective, cost-efficient and reversible therapeutic modality for chronic neuropathic pain resistant to pharmacological treatment. In further studies, the optimal frequency of administered current should be established to maximize the effectiveness of the SCS technique.

5.2.2. Dorsal Root Ganglion Stimulation

Dorsal root ganglion stimulation (DRGS) is a novel, more focused neuromodulatory technique for the treatment of neuropathic pain syndromes compared with standard SRS, enabling the inhibition of pain with even sub-dermatomal accuracy [207]. Moreover, DRGS may require less energy for stimulation and induce postural-independent paresthesia due to the lack of a CSF layer between electrodes and the stimulated dorsal root ganglion. On the other hand, the CSF layer present between SCS electrodes and nerve fibers results in an urgent need for higher current intensity and variable intensity of paresthesia due to the position-dependent thickness of the CSF layer [207]. Contrastingly, the implantation of a DRGS device is more complicated than SCS implantation. Regarding the site of DRGS lead implantation, the T12 nerve root seems to be an optimal location to treat low back pain [208]. DRGS has demonstrated beneficial outcomes in the treatment of discogenic low back pain, failed back surgery syndrome, phantom limb pain, complex regional pain syndrome and postherniorrhaphy groin pain [190,191,192,193,194].
In a multicenter randomized comparative trial, ACCURATE, the use of DRGS showed better outcomes in pain relief and resulted in less postural-dependent paresthesia compared with SCS in 152 patients diagnosed with complex regional pain syndrome and causalgia [209]. A currently recruiting BOOST-DRG study will compare the effectiveness of SCS, DRGS and a combined approach in the treatment of refractory chronic lower limb neuropathic pain and low back neuropathic pain (NCT04852107).
However, the long-term results of DRGS application for neuropathic pain and its cost-effectiveness remain unknown. Nevertheless, DRGS represents another promising method for the alleviation of drug-resistant neuropathic pain.

5.2.3. Non-Invasive Brain Stimulation

Non-invasive brain stimulation (NIBS) techniques, such as transcranial direct current stimulation (tDCS) and repetitive transcranial magnetic stimulation (rTMS), commonly used in the treatment of psychiatric disorders, have recently been proposed for the management of neuropathic pain. In the vast majority of studies on NIBS, the primary motor cortex area (M1) constitutes the anatomical target for attenuating neuropathic pain. However, non-motor brain areas, such as the dorsolateral prefrontal cortex (DLPFC), premotor cortex (PMC), secondary somatosensory cortex (S2) and frontal cortex, have also been demonstrated as promising neurostimulation targets [210]. In both methods, the motor cortex is transcranially stimulated by an electric current, which may be induced by magnetic stimulation in the case of rTMS or by electric stimulation (1–2 mA) in the case of tDCS [211]. The procedure induces the release of striatal dopamine in the brain cortex, which may contribute to the analgesic effect of NIBS [196]. NIBS is generally considered a well-tolerated method, although some mild complications, such as local headache, tinnitus, burning, and in some cases, seizures, have been observed [197]. Moreover, in patients with aneurysm clips, cochlear implants, deep brain electrodes, epilepsy history and cardiac stimulators, rTMS should not be used [1].
rTMS represents the only NIBS technique approved for clinical use by the Federal Drug Administration with indications for drug-resistant major depression [212,213]. Numerous studies have proven effectiveness of rTMS targeting the M1 area of the motor cortex, also in the treatment of neuropathic pain with a longstanding analgesic effect [214,215,216,217]. A significant improvement in pain intensity has been observed in patients with chronic low back pain treated with rTMS without severe complications, compared with patients treated with physiotherapy and a sham group [218]. A currently recruiting randomized controlled trial will evaluate the effectiveness of rTMS alone and in combination with motor control exercises in relieving chronic low back pain (NCT04555278).
One tDCS technique has shown beneficial outcomes in alleviating neuropathic pain associated with traumatic spinal cord injury, stroke, multiple sclerosis, fibromyalgia, trigeminal neuralgia and diabetes [198]. A randomized controlled trial conducted by Straudi et al. showed significantly reduced pain and increased psychological well-being after the use of tDCS in patients with chronic low back pain compared with a sham-tDCS group [219]. However, a recent meta-analysis did not support these findings and demonstrated non-significant pain reduction after tDCS in patients with non-specific chronic low back pain [220]. A recently completed randomized controlled trial, conducted on 60 patients with chronic non-specific low back pain, evaluated the combined use of tDCS and therapeutic exercise (NCT03503422). However, its results have not been posted.
Nevertheless, NIBS techniques represent a non-invasive and potentially effective therapeutic option for neuropathic pain. However, the exact analgesic mechanisms of NIBS should be investigated in further studies, and multicenter randomized studies should be conducted to enhance clinical evidence for the effectiveness of NIBS [197].

5.2.4. Optogenetic Stimulation

Optogenetic stimulation is a relatively new yet promising therapeutic method that involves affecting (both inhibiting and activating) proteins with light. Specific wavelengths affect light-sensitive transmembrane channels, changing their functioning. One of the regions involved in neuropathic pain is the anterior cingulate cortex (ACC), which is located in the limbic system; however, its detailed role remains unclear [216]. The optogenetic activation of dopamine receptors D1 and D2 in ACC may alleviate pain sensations [217].

5.3. Emerging Therapeutic Approaches

5.3.1. Cannabis-Based Medicines

Recently, cannabis-based medicines have been proposed as a therapeutic option for neuropathic pain due to their potential analgesic properties. However, the evidence for the effectiveness of cannabis-based medicines in reducing the chronic neuropathic pain presented by a Cochrane systematic review was of a very low quality [133]. Moreover, most studies investigating cannabinoids in the management of neuropathic pain are characterized by a short follow-up [132]. Thus, long-term studies are indicated to further evaluate the effects of medical cannabis.
The most common adverse effects are transient and mild, and they include somnolence, confusion, dizziness, headaches, dry mouth, diarrhea, constipation and impaired neurocognitive performance [132]. In rare cases, acute psychosis has been observed [134]. Numerous studies have demonstrated that higher THC concentrations more often induce adverse effects than lower concentrations [221,222], whereas formulations with lower THC doses appear to be non-inferior compared with formulations with higher THC concentrations. Therefore, the use of lower THC concentrations (about 1.29%) is sufficient to obtain the analgesic properties in the treatment of neuropathic pain, and suggested to minimize adverse effects.
Nevertheless, further studies are necessary to precisely evaluate the potential advantages and disadvantages of medical cannabis in the treatment of neuropathic pain.

5.3.2. Stem Cell Therapy

Stem cell transplantation (SCT), as a neuroregenerative therapy, has demonstrated the potential capability to regenerate nervous system injuries, such as spinal cord injury, multiple sclerosis and neurodegenerative disorders [223,224,225,226]. Recent studies indicated that SCT may also be an alternative therapeutic modality for neuropathic pain syndromes [200].
SCT can potentially alleviate neuropathic pain through anti-inflammatory action, the suppression of central sensitization and the inhibition of glial cell activation [200]. In numerous studies on animal models, the potential effectiveness of SCT for neuropathic pain has been proven [227,228,229]. A clinical trial by Vaquero et al. demonstrated that therapy with intrathecally administered mesenchymal stromal cells (MSCs) significantly and progressively decreased neuropathic pain in patients after spinal cord injury (p < 0.003) [199]. Moreover, intradiscally injected stem cells have demonstrated beneficial outcomes in the treatment of discogenic low back pain in several clinical studies [230,231,232]. However, the exact SCT mechanism of action remains unclear and should be explored in further preclinical studies. More research is needed to verify the effectiveness of SCT in relieving neuropathic pain and discogenic low back pain.

5.3.3. Targeted Anti-Inflammatory Agents

Recent evidence indicates that inflammation significantly contributes to the pathomechanism of disorders causing neuropathic pain [233] In this phenomenon, numerous inflammatory cells and regulatory molecules are involved, including cytokines, non-coding RNAs, macrophages, T cells, cytokines and chemokines [233]. Therefore, searching for novel therapeutic agents targeting the inflammatory molecules contributing to the neuropathic pain mechanism is relevant.
Numerous anti-inflammatory agents, such as TNF-α, IL-1β, IL-6 and IL-17 inhibitors, have been tested for this purpose with variable outcomes [234] The increasing number of studies suggests an important regulatory role of non-coding RNAs, such as microRNAs or circRNAs, in the neuroinflammatory processes. This finding creates opportunity for their utilization as therapeutic targets or novel biomarkers for monitoring the effectiveness of neuropathic pain treatment [235]. However, the complexity of the inflammatory microenvironment may hamper the effectiveness of immune-targeted therapeutic strategies. Thus, a deep understanding of molecular patterns in disorders associated with neuropathic pain is necessary to obtain successful outcomes.
Nevertheless, targeted anti-inflammatory agents represent a promising therapeutic option for neuropathic pain syndromes.

5.3.4. Other Emerging Pharmacological Agents

While non-pharmacological methods of neuropathic pain treatment have been increasingly investigated in recent years, novel medicaments have also been extensively studied in preclinical or early clinical studies.
Angiotensin II type 2 receptors (ATR2) are expressed in human nociceptive sensory neurons and may contribute in neuropathic pain modulation [236]. The inhibition of ATR2 may result in analgesic effects, which has been confirmed by increasing clinical evidence regarding the effectiveness of ATR2 inhibitors in the management of neuropathic pain [237,238]. However, recent randomized controlled trials evaluating selective ATR2 inhibitor EMA401 in postherpetic neuralgia and painful diabetic neuropathy (NCT03094195 and NCT03297294) were prematurely terminated due to preclinical evidence of long-term hepatotoxicity [136].
NaV1.7 channels, a subtype of selective sodium channels expressed within the nociceptive neurons, may also contribute to neuropathic pain mechanisms [1]. The application of NaV1.7 antagonists such as TV-45070 and BIIB074 did not significantly decrease pain intensity in randomized controlled trials regarding their effectiveness in postherpetic neuralgia [137,138]. However, NaV1.7 needs further investigation for more clear evidence.
Other potentially effective pharmacological agents for the treatment of neuropathic pain include nerve growth antagonists, oxcarbazepine, flavonoids, selective serotonin reuptake inhibitors (SSRIs) and NMDA antagonists [239,240,241,242,243,244] Despite unclear or poor results of initial research concerning these agents, their therapeutic potential for neuropathic pain should be deeply evaluated in future studies.

6. Conclusions

Available epidemiological studies have demonstrated that a neuropathic component of pain in spine diseases constitutes a common phenomenon. The response of neuropathic pain to conventionally used analgesic drugs such as NSAIDs and opioids is limited. Therefore, the diagnosis of neuropathic pain in diseases of the spine plays a crucial role in the selection of effective therapy and contributes to better clinical outcomes. However, in the diagnosis of patients with spinal disorders, distinguishing between chronic nociceptive pain and neuropathic pain very often remains a challenge. Detailed anamnesis and medical examination constitute crucial factors in increasing the chance of a correct diagnosis. Moreover, various scales and questionnaires, including LANSS, NPQ, DN4 and painDETECT, have been developed to help assess pain in patients and diagnose neuropathic pain correctly.
A difficult diagnostic procedure, methodological variations in existing studies, and as yet not fully understood pathomechanisms of neuropathic pain mean that an effective therapeutic approach or scheme has not been established to date. French guidelines and recommendations of The Special Interest Group on Neuropathic Pain (NeuPSIG) may be useful tools for the management of neuropathic pain caused by spine diseases in clinical practice. According to current guidelines, pharmacotherapy remains a mainstay in first-line treatment for neuropathic pain. However, the mediocre long-term efficacy of pharmacological agents, such as SNRIs, gabapentinoids or TCAs, has led to the development of numerous novel non-pharmacologic approaches. These interventions include SCS, DRGS, NIBS and stem cell therapy. Moreover, novel pharmacologic agents have been extensively studied in recent years, including cannabinoids, ATR2 inhibitors and NaV1.7 inhibitors. However, most of them require further studies before being introduced into clinical practice. Therefore, to date, pharmacotherapy and spinal cord stimulation recommended by recent guidelines remain the most applicable methods of treatment. In the future, to facilitate the diagnosis and management of neuropathic pain, it is essential to identify precise diagnostic criteria and find methods that are less invasive and equally effective as a current treatment, as well as to avoid long-term complications.

Author Contributions

Conceptualization, J.B. and J.L.; methodology, J.B. and M.K.; resources, W.C.; data curation, W.C.; writing—original draft preparation, J.B., M.S., J.L. and M.K.; writing—review and editing, J.B., M.S., J.L. and M.K.; visualization, M.S.; supervision, J.B. and P.K.; project administration, P.K.; funding acquisition, P.K. All authors have read and agreed to the published version of the manuscript.

Funding

This publication was funded by account sources of Medical University of Lublin.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Colloca, L.; Ludman, T.; Bouhassira, D.; Baron, R.; Dickenson, A.H.; Yarnitsky, D.; Freeman, R.; Truini, A.; Attal, N.; Finnerup, N.B.; et al. Neuropathic pain. Nat. Rev. Dis. Prim. 2017, 3, 17002. [Google Scholar] [CrossRef]
  2. Finnerup, N.B.; Kuner, R.; Jensen, T.S. Neuropathic Pain: From Mechanisms to Treatment. Physiol. Rev. 2021, 101, 259–301. [Google Scholar] [CrossRef]
  3. Finnerup, N.B.; Haroutounian, S.; Kamerman, P.; Baron, R.; Bennett, D.L.H.; Bouhassira, D.; Cruccu, G.; Freeman, R.; Hansson, P.; Nurmikko, T.; et al. Neuropathic pain: An updated grading system for research and clinical practice. Pain 2016, 157, 1599. [Google Scholar] [CrossRef]
  4. Murnion, B.P. Neuropathic pain: Current definition and review of drug treatment. Aust. Prescr. 2018, 41, 60. [Google Scholar] [CrossRef]
  5. Jensen, T.S.; Finnerup, N.B. Allodynia and hyperalgesia in neuropathic pain: Clinical manifestations and mechanisms. Lancet. Neurol. 2014, 13, 924–935. [Google Scholar] [CrossRef]
  6. Meacham, K.; Shepherd, A.; Mohapatra, D.P.; Haroutounian, S. Neuropathic Pain: Central vs. Peripheral Mechanisms. Curr. Pain Headache Rep. 2017, 21, 28. [Google Scholar] [CrossRef]
  7. Burke, D.; Fullen, B.M.; Stokes, D.; Lennon, O. Neuropathic pain prevalence following spinal cord injury: A systematic review and meta-analysis. Eur. J. Pain 2017, 21, 29–44. [Google Scholar] [CrossRef]
  8. Miclescu, A.; Straatmann, A.; Gkatziani, P.; Butler, S.; Karlsten, R.; Gordh, T. Chronic neuropathic pain after traumatic peripheral nerve injuries in the upper extremity: Prevalence, demographic and surgical determinants, impact on health and on pain medication. Scand. J. Pain 2019, 20, 95–108. [Google Scholar] [CrossRef]
  9. Pasero, C. Pathophysiology of neuropathic pain. Pain Manag. Nurs. 2004, 5, 3–8. [Google Scholar] [CrossRef]
  10. Litak, J.; Szymoniuk, M.; Czyżewski, W.; Hoffman, Z.; Litak, J.; Sakwa, L.; Kamieniak, P. Metallic Implants Used in Lumbar Interbody Fusion. Materials 2022, 15, 3650. [Google Scholar] [CrossRef]
  11. Litak, J.; Czyżewski, W.; Szymoniuk, M.; Sakwa, L.; Pasierb, B.; Litak, J.; Hoffman, Z.; Kamieniak, P.; Roliński, J. Biological and Clinical Aspects of Metastatic Spinal Tumors. Cancers 2022, 14, 4599. [Google Scholar] [CrossRef] [PubMed]
  12. Litak, J.; Czyzewski, W.; Szymoniuk, M.; Pastuszak, B.; Litak, J.; Litak, G.; Grochowski, C.; Rahnama-Hezavah, M.; Kamieniak, P. Hydroxyapatite Use in Spine Surgery&mdash;Molecular and Clinical Aspect. Materials 2022, 15, 2906. [Google Scholar] [CrossRef] [PubMed]
  13. Yamashita, T.; Takahashi, K.; Yonenobu, K.; Kikuchi, S.I. Prevalence of neuropathic pain in cases with chronic pain related to spinal disorders. J. Orthop. Sci. 2014, 19, 15–21. [Google Scholar] [CrossRef] [PubMed]
  14. El Sissi, W.; Arnaout, A.; Chaarani, M.W.; Fouad, M.; El Assuity, W.; Zalzala, M.; Dershaby, Y.; Youseif, E. Prevalence of neuropathic pain among patients with chronic low-back pain in the Arabian Gulf Region assessed using the leeds assessment of neuropathic symptoms and signs pain scale. J. Int. Med. Res. 2010, 38, 2135–2145. [Google Scholar] [CrossRef]
  15. Attal, N.; Bouhassira, D. Translational neuropathic pain research. Pain 2019, 160, S23–S28. [Google Scholar] [CrossRef]
  16. Magrinelli, F.; Zanette, G.; Tamburin, S. Neuropathic pain: Diagnosis and treatment. Pract. Neurol. 2013, 13, 292–307. [Google Scholar] [CrossRef]
  17. Campbell, J.N.; Meyer, R.A. Mechanisms of Neuropathic Pain. Neuron 2006, 52, 77. [Google Scholar] [CrossRef]
  18. Chul Han, H.; Hyun Lee, D.; Mo Chung, J. Characteristics of ectopic discharges in a rat neuropathic pain model. Pain 2000, 84, 253–261. [Google Scholar] [CrossRef]
  19. Yam, M.F.; Loh, Y.C.; Tan, C.S.; Adam, S.K.; Manan, N.A.; Basir, R. General Pathways of Pain Sensation and the Major Neurotransmitters Involved in Pain Regulation. Int. J. Mol. Sci. 2018, 19, 2164. [Google Scholar] [CrossRef]
  20. Devor, M. Ectopic discharge in Abeta afferents as a source of neuropathic pain. Exp. Brain Res. 2009, 196, 115–128. [Google Scholar] [CrossRef]
  21. Nassar, M.A.; Baker, M.D.; Levato, A.; Ingram, R.; Mallucci, G.; McMahon, S.B.; Wood, J.N. Nerve injury induces robust allodynia and ectopic discharges in Nav1.3 null mutant mice. Mol. Pain 2006, 2, 33. [Google Scholar] [CrossRef]
  22. Costigan, M.; Scholz, J.; Woolf, C.J. Neuropathic Pain: A Maladaptive Response of the Nervous System to Damage. Annu. Rev. Neurosci. 2009, 32, 1. [Google Scholar] [CrossRef] [PubMed]
  23. Latremoliere, A.; Woolf, C.J. Central Sensitization: A Generator of Pain Hypersensitivity by Central Neural Plasticity. J. Pain 2009, 10, 895. [Google Scholar] [CrossRef] [PubMed]
  24. Bhangoo, S.; Ren, D.; Miller, R.J.; Henry, K.J.; Lineswala, J.; Hamdouchi, C.; Li, B.; Monahan, P.E.; Chan, D.M.; Ripsch, M.S.; et al. Delayed functional expression of neuronal chemokine receptors following focal nerve demyelination in the rat: A mechanism for the development of chronic sensitization of peripheral nociceptors. Mol. Pain 2007, 3, 38. [Google Scholar] [CrossRef] [PubMed]
  25. Kemp, A.; Manahan-Vaughan, D. Hippocampal long-term depression and long-term potentiation encode different aspects of novelty accquisition. Proc. Natl. Acad. Sci. USA 2004, 101, 8192–8197. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, J.H.; Wu, C.; Lian, Y.N.; Liu, L.; Li, X.Y. Targeting long-term depression of excitatory synaptic transmission for the treatment of neuropathic pain. FEBS J. 2021, 289, 7334–7342. [Google Scholar] [CrossRef]
  27. Bazzari, A.H.; Bazzari, F.H. Advances in targeting central sensitization and brain plasticity in chronic pain. Egypt. J. Neurol. Psychiatry Neurosurg. 2022, 58, 1–13. [Google Scholar] [CrossRef]
  28. Kopach, O.; Voitenko, N. Spinal AMPA receptors: Amenable players in central sensitization for chronic pain therapy? Channels 2021, 15, 284. [Google Scholar] [CrossRef]
  29. Li, C.; Lei, Y.; Tian, Y.; Xu, S.; Shen, X.; Wu, H.; Bao, S.; Wang, F. The etiological contribution of GABAergic plasticity to the pathogenesis of neuropathic pain. Mol. Pain 2019, 15, 1744806919847366. [Google Scholar] [CrossRef]
  30. Martini, F.J.; Guillamón-Vivancos, T.; Moreno-Juan, V.; Valdeolmillos, M.; López-Bendito, G. Spontaneous Activity in Developing Thalamic and Cortical Sensory Networks. Neuron 2021, 109, 2519. [Google Scholar] [CrossRef]
  31. Cain, S.M.; Garcia, E.; Waheed, Z.; Jones, K.L.; Bushell, T.J.; Snutch, T.P. GABAB receptors suppress burst-firing in reticular thalamic neurons. Channels 2017, 11, 574–586. [Google Scholar] [CrossRef] [PubMed]
  32. Song, Z.; Ansah, O.B.; Meyerson, B.A.; Pertovaara, A.; Linderoth, B. The rostroventromedial medulla is engaged in the effects of spinal cord stimulation in a rodent model of neuropathic pain. Neuroscience 2013, 247, 134–144. [Google Scholar] [CrossRef] [PubMed]
  33. Staud, R. The important role of CNS facilitation and inhibition for chronic pain. Int. J. Clin. Rheumtol. 2013, 8, 639. [Google Scholar] [CrossRef] [PubMed]
  34. Ossipov, M.H.; Dussor, G.O.; Porreca, F. Central modulation of pain. J. Clin. Investig. 2010, 120, 3779. [Google Scholar] [CrossRef]
  35. Suzuki, R.; Kontinen, V.K.; Matthews, E.; Williams, E.; Dickenson, A.H. Enlargement of the receptive field size to low intensity mechanical stimulation in the rat spinal nerve ligation model of neuropathy. Exp. Neurol. 2000, 163, 408–413. [Google Scholar] [CrossRef] [PubMed]
  36. Bouhassira, D. Neuropathic pain: Definition, assessment and epidemiology. Rev. Neurol. 2019, 175, 16–25. [Google Scholar] [CrossRef]
  37. Li, X.; Guo, Q.; Ye, Z.; Wang, E.; Zou, W.; Sun, Z.; He, Z.; Zhong, T.; Weng, Y.; Pan, Y. PPAR γ Prevents Neuropathic Pain by Down-Regulating CX3CR1 and Attenuating M1 Activation of Microglia in the Spinal Cord of Rats Using a Sciatic Chronic Constriction Injury Model. Front. Neurosci. 2021, 15, 620525. [Google Scholar] [CrossRef]
  38. Maves, T.J.; Pechman, P.S.; Gebhart, G.F.; Meller, S.T. Possible chemical contribution from chromic gut sutures produces disorders of pain sensation like those seen in man. Pain 1993, 54, 57–69. [Google Scholar] [CrossRef]
  39. Sandkühler, J. Models and mechanisms of hyperalgesia and allodynia. Physiol. Rev. 2009, 89, 707–758. [Google Scholar] [CrossRef]
  40. Garcia, X.; Del Valle, J.; Escribano, E.; Domenech, J.; Queralt, J. Analgesic and antiallodynic effects of antidepressants after infiltration into the rat. Pharmacology 2010, 86, 216–223. [Google Scholar] [CrossRef]
  41. Marchand, F.; Ardid, D.; Chapuy, E.; Alloui, A.; Jourdan, D.; Eschalier, A. Evidence for an involvement of supraspinal delta- and spinal mu-opioid receptors in the antihyperalgesic effect of chronically administered clomipramine in mononeuropathic rats. J. Pharmacol. Exp. Ther. 2003, 307, 268–274. [Google Scholar] [CrossRef]
  42. Moore, R.A.; Derry, S.; Aldington, D.; Cole, P.; Wiffen, P.J. Amitriptyline for neuropathic pain and fibromyalgia in adults. Cochrane Database Syst. Rev. 2012, 12, CD008242. [Google Scholar] [CrossRef] [PubMed]
  43. Wiffen, P.J.; Derry, S.; Bell, R.F.; Rice, A.S.C.; Tölle, T.R.; Phillips, T.; Moore, R.A. Gabapentin for chronic neuropathic pain in adults. Cochrane Database Syst. Rev. 2017, 6, CD007938. [Google Scholar] [CrossRef] [PubMed]
  44. Ye, G.; Lin, C.; Zhang, Y.; Ma, Z.; Chen, Y.; Kong, L.; Yuan, L.; Ma, T. Quercetin Alleviates Neuropathic Pain in the Rat CCI Model by Mediating AMPK/MAPK Pathway. J. Pain Res. 2021, 14, 1289–1301. [Google Scholar] [CrossRef] [PubMed]
  45. Challa, S.R.; Reddy Challa, S. Surgical animal models of neuropathic pain: Pros and Cons. Int. J. Neurosci. 2014, 125, 170–174. [Google Scholar] [CrossRef] [PubMed]
  46. Schäfers, M.; Cain, D. Segmental spinal nerve ligation model of neuropathic pain. Methods Mol. Med. 2004, 99, 155–166. [Google Scholar] [CrossRef]
  47. Hunter, J.C.; Gogas, K.R.; Hedley, L.R.; Jacobson, L.O.; Kassotakis, L.; Thompson, J.; Fontana, D.J. The effect of novel anti-epileptic drugs in rat experimental models of acute and chronic pain. Eur. J. Pharmacol. 1997, 324, 153–160. [Google Scholar] [CrossRef]
  48. Jwa, H.S.; Kim, Y.H.; Lee, J.; Back, S.K.; Park, C.K. Adipose Tissue-Derived Stem Cells Alleviate Cold Allodynia in a Rat Spinal Nerve Ligation Model of Neuropathic Pain. Stem Cells Int. 2020, 2020, 1–7. [Google Scholar] [CrossRef]
  49. Gold, M.S.; Weinreich, D.; Kim, C.S.; Wang, R.; Treanor, J.; Porreca, F.; Lai, J. Redistribution of NaV1.8 in Uninjured Axons Enables Neuropathic Pain. J. Neurosci. 2003, 23, 158. [Google Scholar] [CrossRef]
  50. Liu, Z.Y.; Song, Z.W.; Guo, S.W.; He, J.S.; Wang, S.Y.; Zhu, J.G.; Yang, H.L.; Liu, J.B. CXCL12/CXCR4 signaling contributes to neuropathic pain via central sensitization mechanisms in a rat spinal nerve ligation model. CNS Neurosci. Ther. 2019, 25, 922. [Google Scholar] [CrossRef] [Green Version]
  51. Korah, H.E.; Cheng, K.; Washington, S.M.; Flowers, M.E.; Stratton, H.J.; Patwardhan, A.; Ibrahim, M.M.; Martin, L.F. Partial Sciatic Nerve Ligation: A Mouse Model of Chronic Neuropathic Pain to Study the Antinociceptive Effect of Novel Therapies. J. Vis. Exp. 2022. [Google Scholar] [CrossRef]
  52. Buffon, A.C.; Javornik, M.A.; Heymanns, A.C.; Salm, D.C.; Horewicz, V.V.; Martins, D.F.; Piovezan, A.P. Role of the endocannabinoid system on the antihyperalgesic action of gabapentin in animal model of neuropathic pain induced by partial sciatic nerve ligation. An. Acad. Bras. Cienc. 2020, 92, e20191155. [Google Scholar] [CrossRef] [PubMed]
  53. Ito, H.; Takemura, Y.; Aoki, Y.; Hattori, M.; Horikawa, H.; Yamazaki, M. Analysis of the effects of a tricyclic antidepressant on secondary sleep disturbance induced by chronic pain in a preclinical model. PLoS ONE 2020, 15, e0243325. [Google Scholar] [CrossRef] [PubMed]
  54. Pokkula, S.; Thakur, S.R. Icariin ameliorates partial sciatic nerve ligation induced neuropathic pain in rats: An evidence of in silico and in vivo studies. J. Pharm. Pharmacol. 2021, 73, 874–880. [Google Scholar] [CrossRef]
  55. Richner, M.; Bjerrum, O.J.; Nykjaer, A.; Vaegter, C.B. The spared nerve injury (SNI) model of induced mechanical allodynia in mice. J. Vis. Exp. 2011, 54, e3092. [Google Scholar] [CrossRef]
  56. Guida, F.; De Gregorio, D.; Palazzo, E.; Ricciardi, F.; Boccella, S.; Belardo, C.; Iannotta, M.; Infantino, R.; Formato, F.; Marabese, I.; et al. Behavioral, Biochemical and Electrophysiological Changes in Spared Nerve Injury Model of Neuropathic Pain. Int. J. Mol. Sci. 2020, 21, 3396. [Google Scholar] [CrossRef]
  57. Allchorne, A.J.; Broom, D.C.; Woolf, C.J. Detection of cold pain, cold allodynia and cold hyperalgesia in freely behaving rats. Mol. Pain 2005, 1, 36–39. [Google Scholar] [CrossRef]
  58. Godai, K.; Moriyama, T. Heme oxygenase-1 in the spinal cord plays crucial roles in the analgesic effects of pregabalin and gabapentin in a spared nerve-injury mouse model. Neurosci. Lett. 2022, 767, 136310. [Google Scholar] [CrossRef]
  59. Arsenault, A.; Sawynok, J. Perisurgical amitriptyline produces a preventive effect on afferent hypersensitivity following spared nerve injury. Pain 2009, 146, 308–314. [Google Scholar] [CrossRef]
  60. Zhao, C.; Tall, J.M.; Meyer, R.A.; Raja, S.N. Antiallodynic effects of systemic and intrathecal morphine in the spared nerve injury model of neuropathic pain in rats. Anesthesiology 2004, 100, 905–911. [Google Scholar] [CrossRef]
  61. Decosterd, I.; Woolf, C.J. Spared nerve injury: An animal model of persistent peripheral neuropathic pain. Pain 2000, 87, 149–158. [Google Scholar] [CrossRef] [PubMed]
  62. He, L.; Zhao, W.; Zhang, L.; Ilango, M.; Zhao, N.; Yang, L.; Guan, Z. Modified Spared Nerve Injury Surgery Model of Neuropathic Pain in Mice. J. Vis. Exp. 2022, 2022, e63362. [Google Scholar] [CrossRef]
  63. Pandey, S.; Dvorakova, M.C. Future Perspective of Diabetic Animal Models. Endocr. Metab. Immune Disord. Drug Targets 2020, 20, 25. [Google Scholar] [CrossRef] [PubMed]
  64. Furman, B.L. Streptozotocin-Induced Diabetic Models in Mice and Rats. Curr. Protoc. Pharmacol. 2015, 70, 5.47.1–5.47.20. [Google Scholar] [CrossRef]
  65. Ighodaro, O.M.; Adeosun, A.M.; Akinloye, O.A. Alloxan-induced diabetes, a common model for evaluating the glycemic-control potential of therapeutic compounds and plants extracts in experimental studies. Medicina 2017, 53, 365–374. [Google Scholar] [CrossRef]
  66. Kowluru, R.A. Retinopathy in a Diet-Induced Type 2 Diabetic Rat Model and Role of Epigenetic Modifications. Diabetes 2020, 69, 689–698. [Google Scholar] [CrossRef]
  67. Bram, Y.; Peled, S.; Brahmachari, S.; Harlev, M.; Gazit, E. Active Immunization Against hIAPP Oligomers Ameliorates the Diabetes- Associated Phenotype in a Transgenic Mice Model. Sci. Rep. 2017, 7, 14031. [Google Scholar] [CrossRef]
  68. Li, J.; Chen, X.; Lu, X.; Zhang, C.; Shi, Q.; Feng, L. Pregabalin treatment of peripheral nerve damage in a murine diabetic peripheral neuropathy model. Acta Endocrinol. 2018, 14, 294–299. [Google Scholar] [CrossRef]
  69. Kim, Y.C.; Oh, E.Y.; Kim, S.H.; Lee, M.G. Pharmacokinetics of diclofenac in rat model of diabetes mellitus induced by alloxan or steptozotocin. Biopharm. Drug Dispos. 2006, 27, 85–92. [Google Scholar] [CrossRef]
  70. Qin, Z.; Wang, L.; Li, G.; Qian, X.; Zhang, J.; Guo, Y.; Liu, G. Analysis of the analgesic effects of tricyclic antidepressants on neuropathic pain, diabetic neuropathic pain, and fibromyalgia in rat models. Saudi J. Biol. Sci. 2020, 27, 2485–2490. [Google Scholar] [CrossRef]
  71. Lotfipour, S.; Smith, M.T. Morphine hyposensitivity in streptozotocin-diabetic rats: Reversal by dietary l-arginine treatment. Clin. Exp. Pharmacol. Physiol. 2018, 45, 42–49. [Google Scholar] [CrossRef] [PubMed]
  72. Crul, T.C.; Stolwijk-Swüste, J.M.; Kopsky, D.J.; Visser-Meily, J.M.A.; Post, M.W.M. Neuropathic pain in spinal cord injury: Topical analgesics as a possible treatment. Spinal Cord Ser. Cases 2020, 6, 1–6. [Google Scholar] [CrossRef]
  73. Wilson, S.; Fredericks, D.C.; Safayi, S.; DeVries-Watson, N.A.; Holland, M.T.; Nagel, S.J.; Gillies, G.T.; Howard, M.A. Ovine Hemisection Model of Spinal Cord Injury. J. Investig. Surg. 2021, 34, 380–392. [Google Scholar] [CrossRef]
  74. Jing, N.; Fang, B.; Li, Z.; Tian, A. Exogenous activation of cannabinoid-2 receptor modulates TLR4/MMP9 expression in a spinal cord ischemia reperfusion rat model. J. Neuroinflammation 2020, 17, 1–14. [Google Scholar] [CrossRef]
  75. Chiu, C.-W.; Cheng, H.; Hsieh, S.-L. Contusion Spinal Cord Injury Rat Model. Bio-Protocol 2017, 7, e2337. [Google Scholar] [CrossRef]
  76. Jiang, L.; Hu, J.; He, S.; Zhang, L.; Zhang, Y. Spinal Neuronal NOS Signaling Contributes to Morphine Cardioprotection in Ischemia Reperfusion Injury in Rats. J. Pharmacol. Exp. Ther. 2016, 358, 450–456. [Google Scholar] [CrossRef]
  77. Hao, J.X.; Xu, X.J.; Yu, Y.X.; Seiger, A.; Wiesenfeld-Hallin, Z. Baclofen reverses the hypersensitivity of dorsal horn wide dynamic range neurons to mechanical stimulation after transient spinal cord ischemia; implications for a tonic GABAergic inhibitory control of myelinated fiber input. J. Neurophysiol. 1992, 68, 392–396. [Google Scholar] [CrossRef]
  78. Hao, J.X.; Yu, Y.X.; Seiger, Å.; Wiesenfeld-Hallin, Z. Systemic tocainide relieves mechanical hypersensitivity and normalizes the responses of hyperexcitable dorsal horn wide-dynamic-range neurons after transient spinal cord ischemia in rats. Exp. Brain Res. 1992, 91, 229–235. [Google Scholar] [CrossRef]
  79. Tetik, Ö.; Islamoǧlu, F.; Göncü, T.; Çekirdekçi, A.; Büket, S. Reduction of spinal cord injury with pentobarbital and hypothermia in a rabbit model. Eur. J. Vasc. Endovasc. Surg. 2002, 24, 540–544. [Google Scholar] [CrossRef]
  80. Sirlak, M.; Eryilmaz, S.; Bahadir Inan, M.; Sirin, Y.S.; Besalti, O.; Yazicioglu, L.; Ozcinar, E.; Erdemli, E.; Tasoz, R.; Elhan, A.H.; et al. Effects of carbamazepine on spinal cord ischemia. J. Thorac. Cardiovasc. Surg. 2008, 136, 1038–1043.e4. [Google Scholar] [CrossRef] [Green Version]
  81. Hulsebosch, C.E.; Xu, G.Y.; Perez-Polo, J.R.; Westlund, K.N.; Taylor, C.P.; McAdoo, D.J. Rodent model of chronic central pain after spinal cord contusion injury and effects of gabapentin. J. Neurotrauma 2000, 17, 1205–1217. [Google Scholar] [CrossRef] [PubMed]
  82. Chen, C.H.; Liao, C.H.; Chen, K.C.; Wang, K.L.; Tseng, X.W.; Tsai, W.K.; Chiang, H.S.; Wu, Y.N. B6 Mouse Strain: The Best Fit for LPS-Induced Interstitial Cystitis Model. Int. J. Mol. Sci. 2021, 22, 12053. [Google Scholar] [CrossRef] [PubMed]
  83. Fattori, V.; Franklin, N.S.; Gonzalez-Cano, R.; Peterse, D.; Ghalali, A.; Madrian, E.; Verri, W.A.; Andrews, N.; Woolf, C.J.; Rogers, M.S. Nonsurgical mouse model of endometriosis-associated pain that responds to clinically active drugs. Pain 2020, 161, 1321–1331. [Google Scholar] [CrossRef]
  84. Song, Z.; Jin, C.; Bian, Z.; Liang, C. Extracorporeal shock wave therapy decreases the number of total and degranulated mast cells and alleviates pelvic pain in a rat model of prostatitis. Mol. Cell. Biochem. 2021, 476, 1905–1913. [Google Scholar] [CrossRef]
  85. Luo, J.; Yang, C.; Luo, X.; Yang, Y.; Li, J.; Song, B.; Zhao, J.; Li, L. Chlorogenic acid attenuates cyclophosphamide-induced rat interstitial cystitis. Life Sci. 2020, 254, 117590. [Google Scholar] [CrossRef]
  86. Rudick, C.N.; Chen, M.C.; Mongiu, A.K.; Klumpp, D.J. Organ cross talk modulates pelvic pain. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2007, 293, R1191–R1198. [Google Scholar] [CrossRef]
  87. Spahr, N.; Hodkinson, D.; Jolly, K.; Williams, S.; Howard, M.; Thacker, M. Distinguishing between nociceptive and neuropathic components in chronic low back pain using behavioural evaluation and sensory examination. Musculoskelet. Sci. Pract. 2017, 27, 40. [Google Scholar] [CrossRef] [PubMed]
  88. Baron, R.; Binder, A.; Attal, N.; Casale, R.; Dickenson, A.H.; Treede, R.D. Neuropathic low back pain in clinical practice. Eur. J. Pain 2016, 20, 861–873. [Google Scholar] [CrossRef]
  89. Marchettini, P.; Lacerenza, M.; Mauri, E.; Marangoni, C. Painful Peripheral Neuropathies. Curr. Neuropharmacol. 2006, 4, 175. [Google Scholar] [CrossRef]
  90. Harden, R.N. Chronic neuropathic pain: Mechanisms, diagnosis, and treatment. Neurologist 2005, 11, 111–122. [Google Scholar] [CrossRef]
  91. Attal, N.; Bouhassira, D.; Baron, R. Diagnosis and assessment of neuropathic pain through questionnaires. Lancet Neurol. 2018, 17, 456–466. [Google Scholar] [CrossRef] [PubMed]
  92. Szewczyk, A.K.; Jamroz-Wiśniewska, A.; Rejdak, K. Possible Neuropathic Pain in Clinical Practice—Review on Selected Diagnostic Tools and Its Further Challenges. Diagnostics 2023, 13, 108. [Google Scholar] [CrossRef]
  93. Bouhassira, D.; Lantéri-Minet, M.; Attal, N.; Laurent, B.; Touboul, C. Prevalence of chronic pain with neuropathic characteristics in the general population. Pain 2008, 136, 380–387. [Google Scholar] [CrossRef]
  94. Kim, K.H.; Moon, S.-H.; Hwang, C.-J.; Cho, Y.E. Prevalence of Neuropathic Pain in Patients Scheduled for Lumbar Spine Surgery: Nationwide, Multicenter, Prospective Study. Pain Physician 2015, 18, E889–E897. [Google Scholar]
  95. Park, S.Y.; An, H.S.; Moon, S.H.; Lee, H.M.; Suh, S.W.; Chen, D.; Jeon, J.H. Neuropathic Pain Components in Patients with Lumbar Spinal Stenosis. Yonsei Med. J. 2015, 56, 1044–1050. [Google Scholar] [CrossRef] [PubMed]
  96. Hassan, A.E.; Saleh, H.A.; Baroudy, Y.M.; Abdul-Rahman, K.I.; Najjar, M.W.; Kazi, M.S.; El-Gazar, M.A.; Hafez, M.A.; Abdullah, M.A.; Abdul-Rahman, Y.A.; et al. Prevalence of neuropathic pain among patients suffering from chronic low back pain in Saudi Arabia. Saudi Med. J. 2004, 25, 1986–1990. [Google Scholar] [PubMed]
  97. Attal, N.; Perrot, S.; Fermanian, J.; Bouhassira, D. The neuropathic components of chronic low back pain: A prospective multicenter study using the DN4 Questionnaire. J. Pain 2011, 12, 1080–1087. [Google Scholar] [CrossRef]
  98. Bennett, M.I.; Attal, N.; Backonja, M.M.; Baron, R.; Bouhassira, D.; Freynhagen, R.; Scholz, J.; Tölle, T.R.; Wittchen, H.U.; Jensen, T.S. Using screening tools to identify neuropathic pain. Pain 2007, 127, 199–203. [Google Scholar] [CrossRef]
  99. Moulin, D.E.; Clark, A.J.; Gilron, I.; Ware, M.A.; Watson, C.P.N.; Sessle, B.J.; Coderre, T.; Morley-Forster, P.K.; Stinson, J.; Boulanger, A.; et al. Pharmacological management of chronic neuropathic pain—Consensus statement and guidelines from the Canadian Pain Society. Pain Res. Manag. 2007, 12, 13–21. [Google Scholar] [CrossRef]
  100. Gronseth, G.; Cruccu, G.; Alksne, J.; Argoff, C.; Brainin, M.; Burchiel, K.; Nurmikko, T.; Zakrzewska, J.M. Practice parameter: The diagnostic evaluation and treatment of trigeminal neuralgia (an evidence-based review): Report of the Quality Standards Subcommittee of the American Academy of Neurology and the European Federation of Neurological Societies. Neurology 2008, 71, 1183–1190. [Google Scholar] [CrossRef]
  101. Jensen, T.S.; Madsen, C.S.; Finnerup, N.B. Pharmacology and treatment of neuropathic pains. Curr. Opin. Neurol. 2009, 22, 467–474. [Google Scholar] [CrossRef] [PubMed]
  102. Baron, R.; Freynhagen, R.; Tölle, T.R.; Cloutier, C.; Leon, T.; Murphy, T.K.; Phillips, K. The efficacy and safety of pregabalin in the treatment of neuropathic pain associated with chronic lumbosacral radiculopathy. Pain 2010, 150, 420–427. [Google Scholar] [CrossRef] [PubMed]
  103. Baron, R.; Binder, A.; Wasner, G. Neuropathic pain: Diagnosis, pathophysiological mechanisms, and treatment. Lancet. Neurol. 2010, 9, 807–819. [Google Scholar] [CrossRef] [PubMed]
  104. Moisset, X.; Bouhassira, D.; Attal, N. French guidelines for neuropathic pain: An update and commentary. Rev. Neurol. 2021, 177, 834–837. [Google Scholar] [CrossRef]
  105. Finnerup, N.B.; Attal, N.; Haroutounian, S.; McNicol, E.; Baron, R.; Dworkin, R.H.; Gilron, I.; Haanpää, M.; Hansson, P.; Jensen, T.S.; et al. Pharmacotherapy for neuropathic pain in adults: A systematic review and meta-analysis. Lancet. Neurol. 2015, 14, 162–173. [Google Scholar] [CrossRef]
  106. Varshney, V.; Osborn, J.; Chaturvedi, R.; Shah, V.; Chakravarthy, K. Advances in the interventional management of neuropathic pain. Ann. Transl. Med. 2021, 9, 187. [Google Scholar] [CrossRef]
  107. Ghayur, M.N. Potential Adverse Consequences of Combination Therapy with Gabapentin and Pregabalin. Case Rep. Med. 2021, 2021, 1–4. [Google Scholar] [CrossRef]
  108. Cavalli, E.; Mammana, S.; Nicoletti, F.; Bramanti, P.; Mazzon, E. The neuropathic pain: An overview of the current treatment and future therapeutic approaches. Int. J. Immunopathol. Pharmacol. 2019, 33, 2058738419838383. [Google Scholar] [CrossRef]
  109. Kato, J.; Baba, M.; Kuroha, M.; Kakehi, Y.; Murayama, E.; Wasaki, Y.; Ohwada, S. Safety and Efficacy of Mirogabalin for Peripheral Neuropathic Pain: Pooled Analysis of Two Pivotal Phase III Studies. Clin. Ther. 2021, 43, 822–835.e16. [Google Scholar] [CrossRef]
  110. Akazawa, T.; Inoue, G.; Tanaka, M.; Umehara, T.; Nagai, T.; Oshita, Y.; Imura, T.; Miyagi, M.; Saito, W.; Sako, K.; et al. Somnolence and Dizziness During Mirogabalin Treatment in Patients With Neuropathic Pain Related to Lumbar Disease Who Switched From Pregabalin: A Retrospective Study. Glob. Spine J. 2021. [Google Scholar] [CrossRef]
  111. Obata, H. Analgesic Mechanisms of Antidepressants for Neuropathic Pain. Int. J. Mol. Sci. 2017, 18, 2483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Shinu, P.; Morsy, M.A.; Nair, A.B.; Al Mouslem, A.K.; Venugopala, K.N.; Goyal, M.; Bansal, M.; Jacob, S.; Deb, P.K. Novel Therapies for the Treatment of Neuropathic Pain: Potential and Pitfalls. J. Clin. Med. 2022, 11, 3002. [Google Scholar] [CrossRef] [PubMed]
  113. Szok, D.; Tajti, J.; Nyári, A.; Vécsei, L. Therapeutic Approaches for Peripheral and Central Neuropathic Pain. Behav. Neurol. 2019, 2019, 8685954. [Google Scholar] [CrossRef] [PubMed]
  114. Tzadok, R.; Ablin, J.N. Current and Emerging Pharmacotherapy for Fibromyalgia. Pain Res. Manag. 2020, 2020, 6541798–6541799. [Google Scholar] [CrossRef]
  115. Qureshi, Z.; Ali, M.N.; Khalid, M. An Insight into Potential Pharmacotherapeutic Agents for Painful Diabetic Neuropathy. J. Diabetes Res. 2022, 2022, 1–19. [Google Scholar] [CrossRef]
  116. Pergolizzi, J.V.; Raffa, R.B.; Taylor, R.; Rodriguez, G.; Nalamachu, S.; Langley, P. A review of duloxetine 60 mg once-daily dosing for the management of diabetic peripheral neuropathic pain, fibromyalgia, and chronic musculoskeletal pain due to chronic osteoarthritis pain and low back pain. Pain Pract. 2013, 13, 239–252. [Google Scholar] [CrossRef]
  117. Nudell, Y.; Dym, H.; Sun, F.; Benichou, M.; Malakan, J.; Halpern, L.R. Pharmacologic Management of Neuropathic Pain. Oral Maxillofac. Surg. Clin. North Am. 2022, 34, 61–81. [Google Scholar] [CrossRef]
  118. Voute, M.; Morel, V.; Pickering, G. Topical Lidocaine for Chronic Pain Treatment. Drug Des. Devel. Ther. 2021, 15, 4091. [Google Scholar] [CrossRef]
  119. Attal, N. Pharmacological treatments of neuropathic pain: The latest recommendations. Rev. Neurol. 2019, 175, 46–50. [Google Scholar] [CrossRef]
  120. Hermanns, H.; Hollmann, M.W.; Stevens, M.F.; Lirk, P.; Brandenburger, T.; Piegeler, T.; Werdehausen, R. Molecular mechanisms of action of systemic lidocaine in acute and chronic pain: A narrative review. Br. J. Anaesth. 2019, 123, 335–349. [Google Scholar] [CrossRef]
  121. Derry, S.; Rice, A.S.C.; Cole, P.; Tan, T.; Moore, R.A. Topical capsaicin (high concentration) for chronic neuropathic pain in adults. Cochrane Database Syst. Rev. 2017, 1, CD007393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Abrams, R.M.C.; Pedowitz, E.J.; Simpson, D.M. A critical review of the capsaicin 8% patch for the treatment of neuropathic pain associated with diabetic peripheral neuropathy of the feet in adults. Expert Rev. Neurother. 2021, 21, 259–266. [Google Scholar] [CrossRef]
  123. Zhang, H.; Lian, Y.; Xie, N.; Chen, C.; Zheng, Y. Single-dose botulinum toxin type a compared with repeated-dose for treatment of trigeminal neuralgia: A pilot study. J. Headache Pain 2017, 18, 1–5. [Google Scholar] [CrossRef]
  124. Chen, L.; Zhang, Y.; Chen, Y.; Wang, T.; Sun, K.; Tang, H.; Shen, W.; Ji, F. Efficacy and Safety of Botulinum Toxin A and Pulsed Radiofrequency on Postherpetic Neuralgia: A Randomized Clinical Trial. Contrast Media Mol. Imaging 2022, 2022, 1–9. [Google Scholar] [CrossRef]
  125. Taheri, M.; Sedaghat, M.; Solhpour, A.; Rostami, P.; Safarpour Lima, B. The Effect of Intradermal Botulinum Toxin a injections on painful diabetic polyneuropathy. Diabetes Metab. Syndr. 2020, 14, 1823–1828. [Google Scholar] [CrossRef] [PubMed]
  126. Chun, A.; Levy, I.; Yang, A.; Delgado, A.; Tsai, C.Y.; Leung, E.; Taylor, K.; Kolakowsky-Hayner, S.; Huang, V.; Escalon, M.; et al. Treatment of at-level spinal cord injury pain with botulinum toxin A. Spinal Cord Ser. Cases 2019, 5, 1–9. [Google Scholar] [CrossRef] [PubMed]
  127. Kocot-Kępska, M.; Zajaczkowska, R.; Mika, J.; Kopsky, D.J.; Wordliczek, J.; Dobrogowski, J.; Przeklasa-Muszyńska, A. Topical Treatments and Their Molecular/Cellular Mechanisms in Patients with Peripheral Neuropathic Pain-Narrative Review. Pharmaceutics 2021, 13, 450. [Google Scholar] [CrossRef]
  128. Sahoo, J.; Jena, D.; Viswanath, A.; Barman, A. Injection Botulinum Toxin A in Treatment of Resistant Chronic Low Back Pain: A Prospective Open-Label Study. Cureus 2021, 13, e17811. [Google Scholar] [CrossRef]
  129. Attal, N.; Bouhassira, D. Advances in the treatment of neuropathic pain. Curr. Opin. Neurol. 2021, 34, 631–637. [Google Scholar] [CrossRef]
  130. Rafiullah, M.; Siddiqui, K. Pharmacological Treatment of Diabetic Peripheral Neuropathy: An Update. CNS Neurol. Disord. Drug Targets 2022, 21, 884–900. [Google Scholar] [CrossRef]
  131. Hodder, S.L.; Feinberg, J.; Strathdee, S.A.; Shoptaw, S.; Altice, F.L.; Ortenzio, L.; Beyrer, C. The opioid crisis and HIV in the USA: Deadly synergies. Lancet 2021, 397, 1139–1150. [Google Scholar] [CrossRef] [PubMed]
  132. Bennici, A.; Mannucci, C.; Calapai, F.; Cardia, L.; Ammendolia, I.; Gangemi, S.; Calapai, G.; Soler, D.G. Safety of Medical Cannabis in Neuropathic Chronic Pain Management. Molecules 2021, 26, 6257. [Google Scholar] [CrossRef] [PubMed]
  133. Mücke, M.; Phillips, T.; Radbruch, L.; Petzke, F.; Häuser, W. Cannabis-based medicines for chronic neuropathic pain in adults. Cochrane Database Syst. Rev. 2018, 3, CD012182. [Google Scholar] [CrossRef] [PubMed]
  134. Pierre, J.M. Psychosis associated with medical marijuana: Risk vs. benefits of medicinal cannabis use. Am. J. Psychiatry 2010, 167, 598–599. [Google Scholar] [CrossRef]
  135. Campos, R.M.P.; Aguiar, A.F.L.; Paes-Colli, Y.; Trindade, P.M.P.; Ferreira, B.K.; de Melo Reis, R.A.; Sampaio, L.S. Cannabinoid Therapeutics in Chronic Neuropathic Pain: From Animal Research to Human Treatment. Front. Physiol. 2021, 12, 2180. [Google Scholar] [CrossRef]
  136. Rice, A.S.C.; Dworkin, R.H.; Finnerup, N.B.; Attal, N.; Anand, P.; Freeman, R.; Piaia, A.; Callegari, F.; Doerr, C.; Mondal, S.; et al. Efficacy and safety of EMA401 in peripheral neuropathic pain: Results of 2 randomised, double-blind, phase 2 studies in patients with postherpetic neuralgia and painful diabetic neuropathy. Pain 2021, 162, 2578–2589. [Google Scholar] [CrossRef]
  137. Zakrzewska, J.M.; Palmer, J.; Morisset, V.; Giblin, G.M.; Obermann, M.; Ettlin, D.A.; Cruccu, G.; Bendtsen, L.; Estacion, M.; Derjean, D.; et al. Safety and efficacy of a Nav1.7 selective sodium channel blocker in patients with trigeminal neuralgia: A double-blind, placebo-controlled, randomised withdrawal phase 2a trial. Lancet. Neurol. 2017, 16, 291–300. [Google Scholar] [CrossRef]
  138. Price, N.; Namdari, R.; Neville, J.; Proctor, K.J.W.; Kaber, S.; Vest, J.; Fetell, M.; Malamut, R.; Sherrington, R.P.; Pimstone, S.N.; et al. Safety and Efficacy of a Topical Sodium Channel Inhibitor (TV-45070) in Patients With Postherpetic Neuralgia (PHN): A Randomized, Controlled, Proof-of-Concept, Crossover Study, With a Subgroup Analysis of the Nav1.7 R1150W Genotype. Clin. J. Pain 2017, 33, 310–318. [Google Scholar] [CrossRef]
  139. Mathieson, S.; Lin, C.W.C.; Underwood, M.; Eldabe, S. Pregabalin and gabapentin for pain. BMJ 2020, 369, m1315. [Google Scholar] [CrossRef]
  140. National Institute for Health and Care Excellence (NICE). Neuropathic Pain in Adults: Pharmacological Management in Non-Specialist Settings; National Institute for Health and Care Excellence (NICE): London, UK, 2020. [Google Scholar]
  141. Moisset, X.; Bouhassira, D.; Avez Couturier, J.; Alchaar, H.; Conradi, S.; Delmotte, M.H.; Lanteri-Minet, M.; Lefaucheur, J.P.; Mick, G.; Piano, V.; et al. Pharmacological and non-pharmacological treatments for neuropathic pain: Systematic review and French recommendations. Rev. Neurol. 2020, 176, 325–352. [Google Scholar] [CrossRef]
  142. Robertson, K.; Marshman, L.A.G.; Plummer, D.; Downs, E. Effect of Gabapentin vs Pregabalin on Pain Intensity in Adults With Chronic Sciatica: A Randomized Clinical Trial. JAMA Neurol. 2019, 76, 28–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Yaksi, A.; Özgönenel, L.; Özgönenel, B. The efficiency of gabapentin therapy in patients with lumbar spinal stenosis. Spine 2007, 32, 939–942. [Google Scholar] [CrossRef] [PubMed]
  144. Kasimcan, O.; Kaptan, H. Efficacy of gabapentin for radiculopathy caused by lumbar spinal stenosis and lumbar disk hernia. Neurol. Med. Chir. 2010, 50, 1070–1073. [Google Scholar] [CrossRef]
  145. Bussières, A.; Cancelliere, C.; Ammendolia, C.; Comer, C.M.; Al Zoubi, F.; Châtillon, C.E.; Chernish, G.; Cox, J.M.; Gliedt, J.A.; Haskett, D.; et al. Non-Surgical Interventions for Lumbar Spinal Stenosis Leading To Neurogenic Claudication: A Clinical Practice Guideline. J. Pain 2021, 22, 1015–1039. [Google Scholar] [CrossRef]
  146. Mathieson, S.; Maher, C.G.; McLachlan, A.J.; Latimer, J.; Koes, B.W.; Hancock, M.J.; Harris, I.; Day, R.O.; Billot, L.; Pik, J.; et al. Trial of Pregabalin for Acute and Chronic Sciatica. N. Engl. J. Med. 2017, 376, 1111–1120. [Google Scholar] [CrossRef] [PubMed]
  147. Ebell, M.H. Pregabalin Does Not Decrease the Pain of Sciatica. Am. Fam. Physician 2017, 96, 260. [Google Scholar]
  148. Robertson, K.; Marshman, L.A.G.; Plummer, D. Pregabalin and gabapentin for the treatment of sciatica. J. Clin. Neurosci. 2016, 26, 1–7. [Google Scholar] [CrossRef]
  149. Giménez-Campos, M.S.; Pimenta-Fermisson-Ramos, P.; Díaz-Cambronero, J.I.; Carbonell-Sanchís, R.; López-Briz, E.; Ruíz-García, V. A systematic review and meta-analysis of the effectiveness and adverse events of gabapentin and pregabalin for sciatica pain. Aten. Primaria 2022, 54, 102144. [Google Scholar] [CrossRef]
  150. Attal, N.; Barrot, M. Is Pregabalin Ineffective in Acute or Chronic Sciatica? N. Engl. J. Med. 2017, 376, 1169–1170. [Google Scholar] [CrossRef]
  151. Deeks, E.D. Mirogabalin: First Global Approval. Drugs 2019, 79, 463–468. [Google Scholar] [CrossRef]
  152. Kim, K.; Isu, T.; Kokubo, R.; Iwamoto, N.; Morimoto, D.; Kawauchi, M.; Morita, A. Therapeutic Effect of Mirogabalin on Peripheral Neuropathic Pain due to Lumbar Spine Disease. Asian Spine J. 2021, 15, 349–356. [Google Scholar] [CrossRef] [PubMed]
  153. Nikaido, T.; Takatsuna, H.; Tabata, S.; Shiosakai, K.; Nakatani, T.; Konno, S. ichi Efficacy and Safety of Add-on Mirogabalin to NSAIDs in Lumbar Spinal Stenosis with Peripheral Neuropathic Pain: A Randomized, Open-Label Study. Pain Ther. 2022, 11, 1195–1214. [Google Scholar] [CrossRef] [PubMed]
  154. Dharmshaktu, P.; Tayal, V.; Kalra, B.S. Efficacy of antidepressants as analgesics: A review. J. Clin. Pharmacol. 2012, 52, 6–17. [Google Scholar] [CrossRef]
  155. Asensi-Cantó, A.; López-Abellán, M.D.; Castillo-Guardiola, V.; Hurtado, A.M.; Martínez-Penella, M.; Luengo-Gil, G.; Conesa-Zamora, P. Antitumoral Effects of Tricyclic Antidepressants: Beyond Neuropathic Pain Treatment. Cancers 2022, 14, 3248. [Google Scholar] [CrossRef] [PubMed]
  156. Marsden, J.; White, M.; Annand, F.; Burkinshaw, P.; Carville, S.; Eastwood, B.; Kelleher, M.; Knight, J.; O’Connor, R.; Tran, A.; et al. Medicines associated with dependence or withdrawal: A mixed-methods public health review and national database study in England. Lancet Psychiatry 2019, 6, 935–950. [Google Scholar] [CrossRef]
  157. Ilmanita, D.; Hidayati, H.B. Tricyclic Antidepressants in Chronic Low Back Pain: A Review. J. Islam. Pharm. 2019, 4, 21–26. [Google Scholar] [CrossRef]
  158. Orbai, A.-M.; Meyerhoff, J.O. The effectiveness of tricyclic antidepressants on lumbar spinal stenosis. Bull. NYU Hosp. Jt. Dis. 2010, 68, 22–24. [Google Scholar]
  159. Ferreira, G.E.; McLachlan, A.J.; Lin, C.W.C.; Zadro, J.R.; Abdel-Shaheed, C.; O’Keeffe, M.; Maher, C.G. Efficacy and safety of antidepressants for the treatment of back pain and osteoarthritis: Systematic review and meta-analysis. BMJ 2021, 372, m4825. [Google Scholar] [CrossRef]
  160. Park, K.; Kim, S.; Ko, Y.J.; Park, B.J. Duloxetine and cardiovascular adverse events: A systematic review and meta-analysis. J. Psychiatr. Res. 2020, 124, 109–114. [Google Scholar] [CrossRef]
  161. Gong, H.; Du, X.; Wu, L.; Wei, J.; Tan, Q.; Zhao, R.; Lei, M.; Zhao, L. Discontinuation Syndrome of Extended-Release Venlafaxine during the COVID-19 Epidemic. Psychiatr Danub 2021, 33, 121–122. [Google Scholar]
  162. Konno, S.; Oda, N.; Ochiai, T.; Alev, L. Randomized, Double-blind, Placebo-controlled Phase III Trial of Duloxetine Monotherapy in Japanese Patients With Chronic Low Back Pain. Spine 2016, 41, 1709–1717. [Google Scholar] [CrossRef] [PubMed]
  163. Cawston, H.; Davie, A.; Paget, M.A.; Skljarevski, V.; Happich, M. Efficacy of duloxetine versus alternative oral therapies: An indirect comparison of randomised clinical trials in chronic low back pain. Eur. Spine J. 2013, 22, 1996–2009. [Google Scholar] [CrossRef] [PubMed]
  164. Schukro, R.P.; Oehmke, M.J.; Geroldinger, A.; Heinze, G.; Kress, H.G.; Pramhas, S. Efficacy of Duloxetine in Chronic Low Back Pain with a Neuropathic Component: A Randomized, Double-blind, Placebo-controlled Crossover Trial. Anesthesiology 2016, 124, 150–158. [Google Scholar] [CrossRef] [PubMed]
  165. Hirase, T.; Hirase, J.; Ling, J.; Kuo, P.H.; Hernandez, G.A.; Giwa, K.; Marco, R. Duloxetine for the Treatment of Chronic Low Back Pain: A Systematic Review of Randomized Placebo-Controlled Trials. Cureus 2021, 13, e15169. [Google Scholar] [CrossRef]
  166. Tsuji, O.; Kosugi, S.; Suzuki, S.; Nori, S.; Nagoshi, N.; Okada, E.; Fujita, N.; Yagi, M.; Nakamura, M.; Matsumoto, M.; et al. Effectiveness of Duloxetine for Postsurgical Chronic Neuropathic Disorders after Spine and Spinal Cord Surgery. Asian Spine J. 2021, 15, 650–658. [Google Scholar] [CrossRef]
  167. Santana, J.A.; Klass, S.; Felix, E.R. The Efficacy, Effectiveness and Safety of 5% Transdermal Lidocaine Patch for Chronic Low Back Pain: A Narrative Review. PMR 2020, 12, 1260–1267. [Google Scholar] [CrossRef]
  168. Hashmi, J.A.; Baliki, M.N.; Huang, L.; Parks, E.L.; Chanda, M.L.; Schnitzer, T.; Apkarian, A.V. Lidocaine patch (5%) is no more potent than placebo in treating chronic back pain when tested in a randomised double blind placebo controlled brain imaging study. Mol. Pain 2012, 8, 29. [Google Scholar] [CrossRef]
  169. Imamura, M.; Imamura, S.T.; Targino, R.A.; Morales-Quezada, L.; Onoda Tomikawa, L.C.; Onoda Tomikawa, L.G.; Alfieri, F.M.; Filippo, T.R.; Da Rocha, I.D.; Neto, R.B.; et al. Paraspinous Lidocaine Injection for Chronic Nonspecific Low Back Pain: A Randomized Controlled Clinical Trial. J. Pain 2016, 17, 569. [Google Scholar] [CrossRef]
  170. Frias, B.; Merighi, A. Capsaicin, Nociception and Pain. Molecules 2016, 21, 797. [Google Scholar] [CrossRef]
  171. Zis, P.; Bernali, N.; Argira, E.; Siafaka, I.; Vadalouca, A. Effectiveness and Impact of Capsaicin 8% Patch on Quality of Life in Patients with Lumbosacral Pain: An Open-label Study. Pain Physician 2016, 19, E1049–E1053. [Google Scholar] [CrossRef]
  172. Baron, R.; Treede, R.D.; Birklein, F.; Cegla, T.; Freynhagen, R.; Heskamp, M.L.; Kern, K.U.; Maier, C.; Rolke, R.; Seddigh, S.; et al. Treatment of painful radiculopathies with capsaicin 8% cutaneous patch. Curr. Med. Res. Opin. 2017, 33, 1401–1411. [Google Scholar] [CrossRef] [PubMed]
  173. Waseem, Z.; Boulias, C.; Gordon, A.; Ismail, F.; Sheean, G.; Furlan, A.D. Botulinum toxin injections for low-back pain and sciatica. Cochrane Database Syst. Rev. 2011, 1, CD008257. [Google Scholar] [CrossRef] [PubMed]
  174. Sloan, G.; Alam, U.; Selvarajah, D.; Tesfaye, S. The Treatment of Painful Diabetic Neuropathy. Curr. Diabetes Rev. 2022, 18, 42–96. [Google Scholar] [CrossRef] [PubMed]
  175. Pergolizzi, J.V.; Taylor, R.; LeQuang, J.A.; Raffa, R.B.; Bisney, J. Tapentadol Extended Release in the Treatment of Severe Chronic Low Back Pain and Osteoarthritis Pain. Pain Ther. 2018, 7, 37–57. [Google Scholar] [CrossRef]
  176. Freo, U.; Furnari, M.; Ambrosio, F.; Navalesi, P. Efficacy and tolerability of tapentadol for the treatment of chronic low back pain in elderly patients. Aging Clin. Exp. Res. 2021, 33, 973–982. [Google Scholar] [CrossRef]
  177. Van de Donk, T.; Van Cosburgh, J.; Van Dasselaar, T.; Van Velzen, M.; Drewes, A.M.; Dahan, A.; Niesters, M. Tapentadol treatment results in long-term pain relief in patients with chronic low back pain and associates with reduced segmental sensitization. Pain Rep. 2020, 5, E877. [Google Scholar] [CrossRef]
  178. Migliorini, F.; Maffulli, N.; Baroncini, A.; Eschweiler, J.; Tingart, M.; Quack, V. Opioids for chronic low back pain management: A Bayesian network meta-analysis. Expert Rev. Clin. Pharmacol. 2021, 14, 635–641. [Google Scholar] [CrossRef]
  179. Henson, J.V.; Varhabhatla, N.C.; Bebic, Z.; Kaye, A.D.; Yong, R.J.; Urman, R.D.; Merkow, J.S. Spinal Cord Stimulation for Painful Diabetic Peripheral Neuropathy: A Systematic Review. Pain Ther. 2021, 10, 895–908. [Google Scholar] [CrossRef]
  180. Moufarrij, N. Epidural hematomas after the implantation of thoracic paddle spinal cord stimulators. J. Neurosurg. 2016, 125, 982–985. [Google Scholar] [CrossRef]
  181. Eldabe, S.; Buchser, E.; Duarte, R.V. Complications of Spinal Cord Stimulation and Peripheral Nerve Stimulation Techniques: A Review of the Literature. Pain Med. 2016, 17, 325–336. [Google Scholar] [CrossRef]
  182. Peeters, J.B.; Raftopoulos, C. Tonic, Burst, High-Density, and 10-kHz High-Frequency Spinal Cord Stimulation: Efficiency and Patients’ Preferences in a Failed Back Surgery Syndrome Predominant Population. Review of Literature. World Neurosurg. 2020, 144, e331–e340. [Google Scholar] [CrossRef] [PubMed]
  183. Lind, G.; Winter, J.; Linderoth, B.; Hellström, P.M. Therapeutic value of spinal cord stimulation in irritable bowel syndrome: A randomized crossover pilot study. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2015, 308, R887–R894. [Google Scholar] [CrossRef]
  184. Mehta, V.; Poply, K.; Ahmad, A.; Lascelles, J.; Elyas, A.; Sharma, S.; Ganeshan, B.; Ellamushi, H.; Nikolic, S. Effectiveness of high dose spinal cord stimulation for non-surgical intractable lumbar radiculopathy—HIDENS Study. Pain Pract. 2022, 22, 233–247. [Google Scholar] [CrossRef] [PubMed]
  185. Hoikkanen, T.; Nissen, M.; Ikäheimo, T.M.; Jyrkkänen, H.K.; Huttunen, J.; Von Und Zu Fraunberg, M. Long-Term Outcome of Spinal Cord Stimulation in Complex Regional Pain Syndrome. Neurosurgery 2021, 89, 597–609. [Google Scholar] [CrossRef] [PubMed]
  186. Mehta, V.; Poply, K.; Ahmad, A.; Lascelles, J.; Elyas, A.; Sharma, S.; Ganeshan, B.; Ellamushi, H.; Nikolic, S. The effect of short-term spinal cord electrical stimulation on patients with postherpetic neuralgia and its effect on sleep quality. Neuro Endocrinol. Lett. 2021, 42, 81–86. [Google Scholar]
  187. Galan, V.; Scowcroft, J.; Chang, P.; Li, S.; Staats, P.; Subbaroyan, J.; Caraway, D. Ten kHz spinal cord stimulation for the treatment of chronic peripheral polyneuropathy: 12-Month results from prospective open-label pilot study. Pain Pract. 2021, 21, 898–906. [Google Scholar] [CrossRef]
  188. Heijmans, L.; Joosten, E.A. Mechanisms and mode of action of spinal cord stimulation in chronic neuropathic pain. Postgrad. Med. 2020, 132, 17–21. [Google Scholar] [CrossRef]
  189. Towers, W.S.; Ding, Y.R.; Kurtom, K.H. Delayed upper extremity paresthesia post permanent implantation of five lead spinal cord stimulator for low back and lower extremity pain: A case report. Clin. Neurol. Neurosurg. 2015, 134, 72–74. [Google Scholar] [CrossRef]
  190. Kallewaard, J.W.; Edelbroek, C.; Terheggen, M.; Raza, A.; Geurts, J.W. A Prospective Study of Dorsal Root Ganglion Stimulation for Non-Operated Discogenic Low Back Pain. Neuromodulation 2020, 23, 196–202. [Google Scholar] [CrossRef]
  191. Chapman, K.B.; Nagrani, S.; Patel, K.V.; Yousef, T.; van Helmond, N. Lumbar Dorsal Root Ganglion Stimulation Lead Placement Using an Outside-In Technique in 4 Patients With Failed Back Surgery Syndrome: A Case Series. AA Pract. 2020, 14, e01300. [Google Scholar] [CrossRef]
  192. Yatziv, S.L.; Devor, M. Suppression of neuropathic pain by selective silencing of dorsal root ganglion ectopia using nonblocking concentrations of lidocaine. Pain 2019, 160, 2105–2114. [Google Scholar] [CrossRef] [PubMed]
  193. Stelter, B.; Karri, J.; Marathe, A.; Abd-Elsayed, A. Dorsal Root Ganglion Stimulation for the Treatment of Non-Complex Regional Pain Syndrome Related Chronic Pain Syndromes: A Systematic Review. Neuromodulation 2021, 24, 622–633. [Google Scholar] [CrossRef] [PubMed]
  194. Schu, S.; Gulve, A.; Eldabe, S.; Baranidharan, G.; Wolf, K.; Demmel, W.; Rasche, D.; Sharma, M.; Klase, D.; Jahnichen, G.; et al. Spinal cord stimulation of the dorsal root ganglion for groin pain-a retrospective review. Pain Pract. 2015, 15, 293–299. [Google Scholar] [CrossRef]
  195. Horan, M.; Jacobsen, A.H.; Scherer, C.; Rosenlund, C.; Gulisano, H.A.; Søe, M.; Sørensen, J.C.H.; Meier, K.; Blichfeldt-Eckhardt, M.R. Complications and Effects of Dorsal Root Ganglion Stimulation in the Treatment of Chronic Neuropathic Pain: A Nationwide Cohort Study in Denmark. Neuromodulation 2021, 24, 729–737. [Google Scholar] [CrossRef]
  196. Aceves-Serrano, L.; Neva, J.L.; Munro, J.; Parent, M.; Boyd, L.A.; Doudet, D.J. Continuous but not intermittent theta burst stimulation decreases striatal dopamine release and cortical excitability. Exp. Neurol. 2022, 354, 389–394. [Google Scholar] [CrossRef]
  197. Zhang, K.L.; Yuan, H.; Wu, F.F.; Pu, X.Y.; Liu, B.Z.; Li, Z.; Li, K.F.; Liu, H.; Yang, Y.; Wang, Y.Y. Analgesic Effect of Noninvasive Brain Stimulation for Neuropathic Pain Patients: A Systematic Review. Pain Ther. 2021, 10, 315. [Google Scholar] [CrossRef] [PubMed]
  198. Yang, S.; Chang, M.C. Transcranial Direct Current Stimulation for the Management of Neuropathic Pain: A Narrative Review. Pain Physician 2021, 24, E771–E781. [Google Scholar] [CrossRef] [PubMed]
  199. Vaquero, J.; Zurita, M.; Rico, M.A.; Aguayo, C.; Fernández, C.; Gutiérrez, R.; Rodríguez-Boto, G.; Saab, A.; Hassan, R.; Ortega, C. Intrathecal administration of autologous bone marrow stromal cells improves neuropathic pain in patients with spinal cord injury. Neurosci. Lett. 2018, 670, 14–18. [Google Scholar] [CrossRef]
  200. Joshi, H.P.; Jo, H.J.; Kim, Y.H.; An, S.B.; Park, C.K.; Han, I. Stem Cell Therapy for Modulating Neuroinflammation in Neuropathic Pain. Int. J. Mol. Sci. 2021, 22, 4853. [Google Scholar] [CrossRef]
  201. Odonkor, C.A.; Orman, S.; Orhurhu, V.; Stone, M.E.; Ahmed, S. Spinal Cord Stimulation vs Conventional Therapies for the Treatment of Chronic Low Back and Leg Pain: A Systematic Review of Health Care Resource Utilization and Outcomes in the Last Decade. Pain Med. 2019, 20, 2479–2494. [Google Scholar] [CrossRef]
  202. Eckermann, J.M.; Pilitsis, J.G.; Vannaboutathong, C.; Wagner, B.J.; Province-Azalde, R.; Bendel, M.A. Systematic Literature Review of Spinal Cord Stimulation in Patients with Chronic Back Pain Without Prior Spine Surgery. Neuromodulation 2021, 25, 648–656. [Google Scholar] [CrossRef]
  203. Dones, I.; Levi, V. Spinal Cord Stimulation for Neuropathic Pain: Current Trends and Future Applications. Brain Sci. 2018, 8, 138. [Google Scholar] [CrossRef] [PubMed]
  204. De Vos, C.C.; Bom, M.J.; Vanneste, S.; Lenders, M.W.P.M.; De Ridder, D. Burst spinal cord stimulation evaluated in patients with failed back surgery syndrome and painful diabetic neuropathy. Neuromodulation 2014, 17, 152–159. [Google Scholar] [CrossRef] [PubMed]
  205. Grider, J.; Manchikanti, L.; Carayannopoulos, A.; Sharma, M.L.; Balog, C.C.; Harned, M.E.; Grami, V.; Justiz, R.; Nouri, K.; Hayek, S.M.; et al. Effectiveness of Spinal Cord Stimulation in Chronic Spinal Pain: A Systematic Review. Pain Physician 2016, 19, E33–E54. [Google Scholar] [CrossRef]
  206. Campwala, Z.; Datta, P.; DiMarzio, M.; Sukul, V.; Feustel, P.J.; Pilitsis, J.G. Spinal Cord Stimulation to Treat Low Back Pain in Patients With and Without Previous Spine Surgery. Neuromodulation 2021, 24, 1363–1369. [Google Scholar] [CrossRef]
  207. Harrison, C.; Epton, S.; Bojanic, S.; Green, A.L.; FitzGerald, J.J. The Efficacy and Safety of Dorsal Root Ganglion Stimulation as a Treatment for Neuropathic Pain: A Literature Review. Neuromodulation Technol. Neural Interface 2018, 21, 225–233. [Google Scholar] [CrossRef]
  208. Chapman, K.B.; Groenen, P.S.; Vissers, K.C.; van Helmond, N.; Stanton-Hicks, M.D. The Pathways and Processes Underlying Spinal Transmission of Low Back Pain: Observations From Dorsal Root Ganglion Stimulation Treatment. Neuromodulation 2021, 24, 610–621. [Google Scholar] [CrossRef]
  209. Deer, T.R.; Levy, R.M.; Kramer, J.; Poree, L.; Amirdelfan, K.; Grigsby, E.; Staats, P.; Burton, A.W.; Burgher, A.H.; Obray, J.; et al. 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 2017, 158, 669–681. [Google Scholar] [CrossRef]
  210. Zang, Y.; Zhang, Y.; Lai, X.; Yang, Y.; Guo, J.; Gu, S.; Zhu, Y. Repetitive Transcranial Magnetic Stimulation for Neuropathic Pain on the Non-Motor Cortex: An Evidence Mapping of Systematic Reviews. Evid. Based. Complement. Alternat. Med. 2021, 2021, 1–16. [Google Scholar] [CrossRef]
  211. Bhattacharya, A.; Mrudula, K.; Sreepada, S.S.; Sathyaprabha, T.N.; Pal, P.K.; Chen, R.; Udupa, K. An Overview of Noninvasive Brain Stimulation: Basic Principles and Clinical Applications. Can. J. Neurol. Sci. 2022, 49, 479–492. [Google Scholar] [CrossRef]
  212. Lefaucheur, J.P.; Aleman, A.; Baeken, C.; Benninger, D.H.; Brunelin, J.; Di Lazzaro, V.; Filipović, S.R.; Grefkes, C.; Hasan, A.; Hummel, F.C.; et al. Evidence-based guidelines on the therapeutic use of repetitive transcranial magnetic stimulation (rTMS): An update (2014–2018). Clin. Neurophysiol. 2020, 131, 474–528. [Google Scholar] [CrossRef] [PubMed]
  213. McClintock, S.M.; Reti, I.M.; Carpenter, L.L.; McDonald, W.M.; Dubin, M.; Taylor, S.F.; Cook, I.A.; O’Reardon, J.; Husain, M.M.; Wall, C.; et al. Consensus Recommendations for the Clinical Application of Repetitive Transcranial Magnetic Stimulation (rTMS) in the Treatment of Depression. J. Clin. Psychiatry 2018, 79, 35–48. [Google Scholar] [CrossRef] [PubMed]
  214. Gatzinsky, K.; Bergh, C.; Liljegren, A.; Silander, H.; Samuelsson, J.; Svanberg, T.; Samuelsson, O. Repetitive transcranial magnetic stimulation of the primary motor cortex in management of chronic neuropathic pain: A systematic review. Scand. J. Pain 2020, 21, 8–21. [Google Scholar] [CrossRef] [PubMed]
  215. Attal, N.; Poindessous-Jazat, F.; De Chauvigny, E.; Quesada, C.; Mhalla, A.; Ayache, S.S.; Fermanian, C.; Nizard, J.; Peyron, R.; Lefaucheur, J.P.; et al. Repetitive transcranial magnetic stimulation for neuropathic pain: A randomized multicentre sham-controlled trial. Brain 2021, 144, 3328–3339. [Google Scholar] [CrossRef]
  216. Attia, M.; McCarthy, D.; Abdelghani, M. Repetitive Transcranial Magnetic Stimulation for Treating Chronic Neuropathic Pain: A Systematic Review. Curr. Pain Headache Rep. 2021, 25, 1–6. [Google Scholar] [CrossRef]
  217. Jiang, X.; Yan, W.; Wan, R.; Lin, Y.; Zhu, X.; Song, G.; Zheng, K.; Wang, Y.; Wang, X. Effects of repetitive transcranial magnetic stimulation on neuropathic pain: A systematic review and meta-analysis. Neurosci. Biobehav. Rev. 2022, 132, 130–141. [Google Scholar] [CrossRef]
  218. Ambriz-Tututi, M.; Alvarado-Reynoso, B.; Drucker-Colín, R. Analgesic effect of repetitive transcranial magnetic stimulation (rTMS) in patients with chronic low back pain. Bioelectromagnetics 2016, 37, 527–535. [Google Scholar] [CrossRef]
  219. Straudi, S.; Buja, S.; Baroni, A.; Pavarelli, C.; Pranovi, G.; Fregni, F.; Basaglia, N. The effects of transcranial direct current stimulation (tDCS) combined with group exercise treatment in subjects with chronic low back pain: A pilot randomized control trial. Clin. Rehabil. 2018, 32, 1348–1356. [Google Scholar] [CrossRef]
  220. Alwardat, M.; Pisani, A.; Etoom, M.; Carpenedo, R.; Chinè, E.; Dauri, M.; Leonardis, F.; Natoli, S. Is transcranial direct current stimulation (tDCS) effective for chronic low back pain? A systematic review and meta-analysis. J. Neural Transm. 2020, 127, 1257–1270. [Google Scholar] [CrossRef]
  221. Wilsey, B.; Marcotte, T.; Deutsch, R.; Gouaux, B.; Sakai, S.; Donaghe, H. Low Dose Vaporized Cannabis Significantly Improves Neuropathic Pain. J. Pain 2013, 14, 136. [Google Scholar] [CrossRef]
  222. Wilsey, B.; Marcotte, T.D.; Deutsch, R.; Zhao, H.; Prasad, H.; Phan, A. An Exploratory Human Laboratory Experiment Evaluating Vaporized Cannabis in the Treatment of Neuropathic Pain from Spinal Cord Injury and Disease. J. Pain 2016, 17, 982. [Google Scholar] [CrossRef] [PubMed]
  223. Szymoniuk, M.; Litak, J.; Sakwa, L.; Dryla, A.; Zezuliński, W.; Czyżewski, W.; Kamieniak, P.; Blicharski, T. Molecular Mechanisms and Clinical Application of Multipotent Stem Cells for Spinal Cord Injury. Cells 2022, 12, 120. [Google Scholar] [CrossRef] [PubMed]
  224. Puranik, N.; Arukha, A.P.; Yadav, S.K.; Yadav, D.; Jin, J.O. Exploring the Role of Stem Cell Therapy in Treating Neurodegenerative Diseases: Challenges and Current Perspectives. Curr. Stem Cell Res. Ther. 2022, 17, 113–125. [Google Scholar] [CrossRef]
  225. Ejma, M.; Madetko, N.; Brzecka, A.; Alster, P.; Budrewicz, S.; Koszewicz, M.; Misiuk-Hojło, M.; Tomilova, I.K.; Somasundaram, S.G.; Kirkland, C.E. The Role of Stem Cells in the Therapy of Stroke. Curr. Neuropharmacol. 2022, 20, 630–647. [Google Scholar] [CrossRef] [PubMed]
  226. Peterson, S.; Jalil, A.; Beard, K.; Kakara, M.; Sriwastava, S. Updates on efficacy and safety outcomes of new and emerging disease modifying therapies and stem cell therapy for Multiple Sclerosis: A review. Mult. Scler. Relat. Disord. 2022, 68, 104125. [Google Scholar] [CrossRef] [PubMed]
  227. Yousof, S.M.; Elsayed, D.A.; El-Baz, A.A.; Sallam, H.S.; Abbas, F. Combined Treatment of Adipose Derived-Mesenchymal Stem Cells and Pregabalin Is Superior to Monotherapy for the Treatment of Neuropathic Pain in Rats. Stem Cells Int. 2021, 2021, 1–9. [Google Scholar] [CrossRef]
  228. Kotb, H.I.; Abedalmohsen, A.M.; Elgamal, A.F.; Mokhtar, D.M.; Abd-Ellatief, R.B. Preemptive Stem Cells Ameliorate Neuropathic Pain in Rats: A Central Component of Preemptive Analgesia. Microsc. Microanal. 2021, 27, 450–456. [Google Scholar] [CrossRef] [PubMed]
  229. Tian, J.; Song, T.; Wang, H.; Wang, W.; Zhang, Z.; Yan, R.; Ma, X.; Hu, Y. Intrathecal Injection of SIRT1-modified Human Mesenchymal Stem Cells Alleviates Neuropathic Pain in Rat. J. Mol. Neurosci. 2021, 71, 972–980. [Google Scholar] [CrossRef]
  230. Urits, I.; Capuco, A.; Sharma, M.; Kaye, A.D.; Viswanath, O.; Cornett, E.M.; Orhurhu, V. Stem Cell Therapies for Treatment of Discogenic Low Back Pain: A Comprehensive Review. Curr. Pain Headache Rep. 2019, 23, 65. [Google Scholar] [CrossRef]
  231. Atluri, S.; Murphy, M.B.; Dragella, R.; Herrera, J.; Boachie-Adjei, K.; Bhati, S.; Manocha, V.; Boddu, N.; Yerramsetty, P.; Syed, Z.; et al. Evaluation of the Effectiveness of Autologous Bone Marrow Mesenchymal Stem Cells in the Treatment of Chronic Low Back Pain Due to Severe Lumbar Spinal Degeneration: A 12-Month, Open-Label, Prospective Controlled Trial. Pain Physician 2022, 25, 193–207. [Google Scholar]
  232. Amirdelfan, K.; Bae, H.; McJunkin, T.; DePalma, M.; Kim, K.; Beckworth, W.J.; Ghiselli, G.; Bainbridge, J.S.; Dryer, R.; Deer, T.R.; et al. Allogeneic mesenchymal precursor cells treatment for chronic low back pain associated with degenerative disc disease: A prospective randomized, placebo-controlled 36-month study of safety and efficacy. Spine J. 2021, 21, 212–230. [Google Scholar] [CrossRef] [PubMed]
  233. Sommer, C.; Leinders, M.; Üçeyler, N. Inflammation in the pathophysiology of neuropathic pain. Pain 2018, 159, 595–602. [Google Scholar] [CrossRef]
  234. Lees, J.G.; Duffy, S.S.; Moalem-Taylor, G. Immunotherapy targeting cytokines in neuropathic pain. Front. Pharmacol. 2013, 4, 142. [Google Scholar] [CrossRef] [PubMed]
  235. Xu, D.; Ma, X.; Sun, C.; Han, J.; Zhou, C.; Chan, M.T.V.; Wu, W.K.K. Emerging roles of circular RNAs in neuropathic pain. Cell Prolif. 2021, 54, e13139. [Google Scholar] [CrossRef]
  236. Balogh, M.; Aguilar, C.; Nguyen, N.T.; Shepherd, A.J. Angiotensin receptors and neuropathic pain. Pain Rep. 2021, 6, e869. [Google Scholar] [CrossRef]
  237. Smith, M.T.; Anand, P.; Rice, A.S.C. Selective small molecule angiotensin II type 2 receptor antagonists for neuropathic pain: Preclinical and clinical studies. Pain 2016, 157, S33–S41. [Google Scholar] [CrossRef] [PubMed]
  238. Rice, A.S.C.; Dworkin, R.H.; McCarthy, T.D.; Anand, P.; Bountra, C.; McCloud, P.I.; Hill, J.; Cutter, G.; Kitson, G.; Desem, N.; et al. EMA401, an orally administered highly selective angiotensin II type 2 receptor antagonist, as a novel treatment for postherpetic neuralgia: A randomised, double-blind, placebo-controlled phase 2 clinical trial. Lancet 2014, 383, 1637–1647. [Google Scholar] [CrossRef]
  239. Wang, H.; Romano, G.; Fedgchin, M.; Russell, L.; Sanga, P.; Kelly, K.M.; Frustaci, M.E.; Thipphawong, J. Fulranumab in Patients With Pain Associated with Postherpetic Neuralgia and Postraumatic Neuropathy: Efficacy, Safety, and Tolerability Results From a Randomized, Double-blind, Placebo-controlled, Phase-2 Study. Clin. J. Pain 2017, 33, 99–108. [Google Scholar] [CrossRef]
  240. Zhou, M.; Chen, N.; He, L.; Yang, M.; Zhu, C.; Wu, F. Oxcarbazepine for neuropathic pain. Cochrane Database Syst. Rev. 2017, 12, CD007963. [Google Scholar] [CrossRef]
  241. Rao, P.N.; Mainkar, O.; Bansal, N.; Rakesh, N.; Haffey, P.; Urits, I.; Orhurhu, V.; Kaye, A.D.; Urman, R.D.; Gulati, A.; et al. Flavonoids in the Treatment of Neuropathic Pain. Curr. Pain Headache Rep. 2021, 25, 1–10. [Google Scholar] [CrossRef]
  242. Saito, H.; Wakai, J.; Sekiguchi, M.; Kikuchi, S.; Konno, S. The effect of selective serotonin reuptake inhibitor (SSRI) on pain-related behavior in a rat model of neuropathic pain. Eur. Spine J. 2014, 23, 2401–2409. [Google Scholar] [CrossRef] [PubMed]
  243. Foley, P.; Parker, R.A.; de Angelis, F.; Connick, P.; Chandran, S.; Young, C.; Weir, C.J.; Chataway, J. Efficacy of Fluoxetine, Riluzole and Amiloride in treating neuropathic pain associated with secondary progressive multiple sclerosis. Pre-specified analysis of the MS-SMART double-blind randomised placebo-controlled trial. Mult. Scler. Relat. Disord. 2022, 63, 103925. [Google Scholar] [CrossRef] [PubMed]
  244. Aiyer, R.; Mehta, N.; Gungor, S.; Gulati, A. A Systematic Review of NMDA Receptor Antagonists for Treatment of Neuropathic Pain in Clinical Practice. Clin. J. Pain 2018, 34, 450–467. [Google Scholar] [CrossRef] [PubMed]
Table 1. Frequency of neuropathic pain in patients with spine disorders investigated by epidemiological studies.
Table 1. Frequency of neuropathic pain in patients with spine disorders investigated by epidemiological studies.
StudySpine DiseaseFrequency of Neuropathic Pain
(Number of Patients)
Kim et al. [94]Overall36.4% (404)
Stenosis with instability31.7% (64)
Stenosis without instability39.5% (219)
Herniated lumbar disc34.7% (119)
Degenerative lumbar scoliosis22.2% (2)
Park et al. [95]Overall36.0% (31)
Radicular pain63.4% (24)
Neurogenic claudication15.6% (7)
Yamashita et al. [13]Overall53.5% (993)
Spondylotic myelopathy77.3%
Ligament ossification75.7%
Low back pain29.4%
Spondylolysis40.4%
Radiculopathy56.9%
El Sissi et al. [14]Overall55% (628)
Table 2. A summary of currently available pharmacological agents for neuropathic pain.
Table 2. A summary of currently available pharmacological agents for neuropathic pain.
Pharmacological AgentMechanism of ActionClinical EffectivenessAdverse EffectsRecommendation by FG [104]References
Pregabalinbinding to the α2-δ subunit of presynaptic voltage-gated Ca2+ channels in the dorsal horn of the spinal cordpostherpetic neuralgia, spinal cord injury, phantom limb syndrome and diabetes-induced neuropathyperipheral edema, somnolence, dizziness, xerostomia and obesitySecond-line treatment[106,107,108]
GabapentinFirst-line treatment
Mirogabalinpostherpetic neuralgia and diabetic neuropathic painsomnolence, dizzinessNot recommended[109,110]
Tricyclic Antidepressantsincreasing the level of noradrenaline in the dorsal horn of the spinal cord and locus coeruleuspostherpetic neuralgia, central postpartum pain, pain after spinal cord injury, and nerve injurycognitive impairment, walking disturbances, urinary retention, constipation, dry mouth, and orthostatic hypotensionFirst-line treatment[105,108,111,112,113]
serotonin and norepinephrine reuptake inhibitorsincreasing the level of noradrenaline in the dorsal horn of the spinal cord and locus coeruelusfibromyalgia, diabetic neuropathy, and low back painlack of appetite, constipation, dry mouth, anxiety, hyperhidrosis and nauseaFirst-line treatment[111,114,115,116,117]
Topical lidocaineblocking voltage-gated Na+ channelspostherpetic neuralgia, postsurgical chronic neuropathic pain and diabetic peripheral neuropathyskin irritationFirst-line treatment[118,119,120]
Capsaicin patchesmodulation of transient receptor potential cation channel vanilloid subfamily member 1 (TRPV1)diabetic peripheral neuropathy, postherpetic neuralgia and HIV neuropathiesskin irritationSecond-line treatment[105,121,122]
Botulinum toxin Amodulation of both peripheral and central sensitizationstrigeminal neuralgia, postherpetic neuralgia, diabetic peripheral neuropathy and pain after spinal cord injurytransient injection site painSecond-line treatment[123,124,125,126,127,128]
Tramadolµ-opioid agonist and 5-HT reuptake inhibitorperipheral neuropathic painSomnolence, confusion, and the risk of misuse and addictionSecond-line treatment[112,129,130,131]
Cannabis-based medicinesbinding to the CB1 and CB2 receptors, COX-2 inhibitionlow-quality evidence for chronic neuropathic painsomnolence, confusion, dizziness, headaches, dry mouth, diarrhea, constipation, impaired neurocognitive performance and psychosisNot recommended[132,133,134,135]
EMA401Angiotensin II type 2 receptor (ATR2) inhibitionpostherpetic neuralgia and painful diabetic neuropathyrisk of long-term hepatotoxicityNot recommended[136]
TV-45070 and BIIB074NaV1.7 inhibitionpostherpetic neuralgialocal skin reactions where application site pain and pruritus (TV-45070); headache, pyrexia, nasopharyngitis, sleep disorder and tremor (BIIB074)Not recommended[137,138]
Table 3. A summary of currently available non-pharmacological therapeutic methods for neuropathic pain.
Table 3. A summary of currently available non-pharmacological therapeutic methods for neuropathic pain.
Therapeutic ApproachMechanism of ActionClinical EffectivenessAdverse EffectsRecommendation by French Guidelines [104]References
Spinal Cord Stimulationinhibition of the C and Aδ-fibers, and an increase in GABA and serotonin levels in the spinal cordperipheral diabetic neuropathy, failed back surgery syndrome, irritable bowel syndrome, painful radiculopathy, complex regional pain syndrome, postherpetic neuralgia and chronic low back painpain at the generator site, infection, nerve injury, epidural hematoma, hardware failuresThird-line treatment[112,179,180,181,182,183,184,185,186,187,188,189]
Dorsal Root Ganglion Stimulationinhibition of the C and Aδ-fibers, and an increase in GABA and serotonin levels in the dorsal horn of the spinal corddiscogenic low back pain, failed back surgery syndrome, phantom limb pain, complex regional pain syndrome and postherniorrhaphy groin painlead fractures, nerve injuryNot recommended[190,191,192,193,194,195]
Repetitive Transcranial Magnetic Stimulationinducing the release of striatal dopamine in the brain cortex through magnetic stimuluspain after spinal cord injury, phantom limb pain, pain secondary to malignancy, postherpetic neuralgia, diabetic neuropathy, chronic low back painheadache, tinnitus, burning and seizuresThird-line treatment[141,196,197]
Transcranial Direct Current Stimulationinducing the release of striatal dopamine in the brain cortex through electric stimulustraumatic spinal cord injury, stroke, multiple sclerosis, fibromyalgia, trigeminal neuralgia and diabetesheadache, tinnitus, burning and seizuresNot recommended[196,197,198]
Stem Cell Therapyanti-inflammatory effect, suppression of central sensitization and inhibition of glial cell activationuncertain evidence for neuropathic pain after spinal cord injuryfever, headache, infections, potential cancerogenicityNot recommended[199,200]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bielewicz, J.; Kamieniak, M.; Szymoniuk, M.; Litak, J.; Czyżewski, W.; Kamieniak, P. Diagnosis and Management of Neuropathic Pain in Spine Diseases. J. Clin. Med. 2023, 12, 1380. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm12041380

AMA Style

Bielewicz J, Kamieniak M, Szymoniuk M, Litak J, Czyżewski W, Kamieniak P. Diagnosis and Management of Neuropathic Pain in Spine Diseases. Journal of Clinical Medicine. 2023; 12(4):1380. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm12041380

Chicago/Turabian Style

Bielewicz, Joanna, Maciej Kamieniak, Michał Szymoniuk, Jakub Litak, Wojciech Czyżewski, and Piotr Kamieniak. 2023. "Diagnosis and Management of Neuropathic Pain in Spine Diseases" Journal of Clinical Medicine 12, no. 4: 1380. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm12041380

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop