Review Article | | Peer-Reviewed

A Chinese Guideline for the Diagnosis and Treatment of Neuropathic Pain

Received: 26 May 2025     Accepted: 13 June 2025     Published: 14 October 2025
Views:       Downloads:
Abstract

Background: Neuropathic pain refers to pain caused by injury or disease affecting the somatosensory nervous system, which is a common and frequently occurring disease in clinical practice, and seriously affects patients' quality of life. However, the treatment of neuropathic pain is a clinical challenge. Objective: To standardize neuropathic pain management, the Pain Medicine Branch of China Association of Health Care for the Elderly convened an expert panel to develop the guideline. Main ideas: Based on high quality evidence-based medical research on the diagnosis and treatment of neuropathic pain published domestically and internationally in the past 10 years, the expert group has formed recommendations for common treatment methods through rigorous argumentation and expert voting, to provide references for standardized diagnosis and treatment of neuropathic pain. This guideline adopts GRADE methodology to evaluate the level of evidence and strength of recommendation for the treatments of common peripheral neuropathic pain and central neuropathic pain. Chinese traditional medicine also plays an important role in the treatment of neuropathic pain, so this guide also provides Chinese traditional medicine drugs and treatment recommendations. Conclusion: The Chinese pain community has proposed the principle of "treatment forward, early intervention, prevention of sensitization, and prevention and treatment of chronic pain", which has played a positive role in improving the clinical diagnosis and treatment level of neuropathic pain.

Published in International Journal of Pain Research (Volume 1, Issue 4)
DOI 10.11648/j.ijpr.20250104.12
Page(s) 56-86
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2025. Published by Science Publishing Group

Keywords

Neuropathic Pain, Postherpetic Neuralgia, Trigeminal Neuralgia, Painful Diabetic Peripheral Neuropathy, Spinal Cord Injury Pain, Guideline

1. Introduction
Neuropathic pain (NP) refers to pain caused by injury or disease affecting the somatosensory nervous system. NP is usually chronic, with persistent or recurrent pain, causing great pain and functional impairment to patients. Various diseases that affect the peripheral or central nervous system, such as neurodegenerative diseases, autoimmune diseases, metabolic diseases, vascular diseases, tumors, infections, poisoning, trauma, genetic diseases, and unexplained neurological disorders, can all induce NP. The prevalence of NP in the general population is 3.0% to 17.0% .
The treatment of NP is full of challenges and difficulties. The Chinese pain community has proposed the principle of "treatment forward, early intervention, prevention of sensitization, and prevention and treatment of chronic pain", which has played a positive role in improving the clinical diagnosis and treatment level of NP. To standardize NP management, the Pain Medicine Branch of China Association of Health Care for the Elderly convened an expert panel to develop the ‘Chinese Guideline for Neuropathic Pain Diagnosis and Treatment’. This evidence-based guideline was formulated through systematic literature reviews of both domestic and international evidence, critical appraisal of expert recommendations through multiple rounds of expert evaluation.
2. Guideline Development Methodology
The literature search period was from January 2014 to June 2024. The search terms included neuropathic pain, peripheral neuropathic pain, central neuropathic pain, etc. Data sourced from Wanfang, China National Knowledge Infrastructure (CNKI), PubMed, and Cochrane Library. Selectted high-grade evidence literatures such as systematic review, meta-analysis, randomized controlled trials (RCTs), expert consensus, and clinical guidelines as the basis. After multiple rounds of discussion and online voting using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) methodology (Table 1 ) and consensus conference method, this guideline was ultimately developed.
Table 1. GRADE System Evidence Quality and Recommendation Strength.

Level

Description

Evidence Quality

High (A)

We are very confident that the true effect lies close to the estimate.

Moderate (B)

We are moderately confident in the effect estimate: The true effect is likely close to the estimate, but there is a possibility of substantial difference.

Low (C)

Our confidence in the effect estimate is limited: The true effect may be substantially different.

Very Low (D)

We have very little confidence in the effect estimate: The true effect is likely substantially different.

Recommendation Strength

Strong (1)

Most individuals in this situation would want the recommended course of action, and only a small proportion would not; most clinicians should adopt the intervention; >70% panel agreement.

Weak (2)

Most individuals would choose the recommended course, but many would not; clinicians should help patients make decisions consistent with values/preferences; 50–70% panel agreement.

No Recommendation (3)

Trade-offs uncertain; target population unclear; insufficient evidence; <50% panel agreement.

3. Pathogenesis
The mechanism of NP is complex and not yet fully understood. Research has shown that pain transmission, modulation pathways, and pathological plasticity changes in cognitive and affective related brain regions caused by injury and disease play an important role . In peripheral sensory nerves, pathological plasticity changes are manifested by abnormal expression of multiple ion channels in dorsal root ganglion and trigeminal ganglion neurons, such as upregulation of sodium and calcium channels, while downregulation of potassium channels, causing abnormal increase in excitability and ectopic discharge, leading to hyperalgesia, hyperalgesia, and spontaneous pain , resulting in plastic changes in pain transmission at all levels of the central nervous system, from the spinal dorsal horn to the sensory cortex. In the early stage of NP, functional changes such as excitatory synaptic transmission efficiency and long term potentiation (LTP) are manifested. In the later stage, the number of excitatory synapses increases while the number of inhibitory synapses decreases, leading to neural circuit remodeling . The plasticity of the pain modulation system changes, leading to a decrease in the function of the descending inhibitory system and an increase in the activity of the descending facilitation system . The functional and structural changes in cognitive and affective related brain regions, such as the hippocampus and prefrontal cortex, lead to cognitive and affective disorders, and the changes in cognitive function enhance pain perception by interfering with the pain modulation system . Research has shown that activation of glial cells and overexpression of inflammatory cytokines such as TNF-α and IL-1β cause neuroinflammation, mediating chronic pain. Inflammatory cytokines regulate the expression of multiple ion channels, causing abnormal elevation of neuronal excitability. By differentially regulating synaptic plasticity in different brain regions, such as upregulating and downregulating the number of excitatory synapses in the spinal dorsal horn and hippocampus, chronic pain and cognitive or affective disorders can be caused. The destruction of the blood-brain barrier and blood-brain barrier caused by peripheral and central nervous system injury, leading to the infiltration of peripheral immune cells into peripheral nerves and brain tissue, is an important trigger of neuroinflammation .
4. Clinical Characteristics of NP
4.1. Peripheral Neuropathic Pain
4.1.1. Trigeminal Neuralgia
(i). Overview
Trigeminal neuralgia (TN) is divided into primary TN and secondary TN. Primary TN is the most common type in clinical practice, and is further divided into classic TN (with neurovascular compression) and idiopathic TN (with no underlying cause identified). Secondary TN (with potential pathology), also known as symptomatic TN, refers to TN caused by secondary damage to the trigeminal nerve caused by various organic diseases inside and outside the skull. The prevalence rate of TN is 182/100,000, and the annual incidence rate is 3-5/100,000, mostly in adults and the elderly .
(ii). Clinical Characteristics
Paroxysmal severe pain occurring in the trigeminal nerve distribution area, often accompanied by electric shock or needle puncture sensation, lasting for several seconds to minutes, with periodic pain and asymptomatic intervals. Pain is often triggered by touching the trigger point and is more common on one side, particularly in the maxillary and mandibular branches. When the pain is severe, it may be accompanied by ipsilateral muscle twitching, facial flushing, tears, and drooling.
(iii). Diagnosis
Typical primary TN is diagnosed based on the third edition of International Classification of Headache Disorders (ICHD-3).
Trigeminal nerve attacks occur in one or more branches, without external radiating pain of the trigeminal nerve.
Include at least 3 of the following 4 items: Intermittent pain lasting for 1-2 minutes; Severe pain; Electric-shock-like pain, lightning-like pain, and lancinating pain; Trigger pain on the affected side.
No clinically significant neurological deficits.
Exclude other diagnoses listed in ICHD-3.
Must fulfill both criteria (i) and (ii), with documentation of ≥3 ipsilateral episodes.
(iv). Differential Diagnosis
TN needs to be differentiated from diseases such as glossopharyngeal neuralgia, toothache, cluster headache, atypical facial pain, sphenopalatine neuralgia, postherpetic neuralgia in the trigeminal nerve area, temporomandibular joint dysfunction syndrome, painful ophthalmoplegia syndrome, coronary heart disease, etc.
4.1.2. Postherpetic Neuralgia
(i). Overview
Postherpetic neuralgia (PHN) is defined as persistent pain lasting for ≥1 month after the resolution of herpes zoster rash, representing the most common complication of herpes zoster . PHN is more common in the elderly, and the incidence increases with age.
(ii). Clinical Characteristics
The most common areas of PHN are the chest and back, followed by the head, face, neck, and lumbar back, with the lowest incidence in the lumbar and sacral regions. The range of pain in patients often coincides or expands with the herpes area. Patients with PHN typically experience persistent baseline pain with paroxysmal worsening. The pain manifests in various neuropathic qualities, including electric-shock-like, lancinating, burning, or stabbing sensations. Frequent occurrences of spontaneous pain, hyperalgesia, hyperalgesia, and abnormal sensations.
PHN patients often have accompanying symptoms such as anxiety, depression, poor sleep, and decreased quality of life.
(iii). Diagnosis and Differential Diagnosis
The diagnosis of PHN is primarily based on patient history and clinical presentation, typically requiring no additional specialized investigations. PHN needs to be differentiated from primary TN, glossopharyngeal neuralgia, occipital neuralgia, intercostal neuralgia, and pain caused by primary tumor compression.
4.1.3. Painful Polyneuropathy
(i). Overview
Painful polyneuropathy refers to a kind of disease with pain symptoms caused by many reasons, such as aberrant metabolism, autoimmune response, family inheritance, infectious diseases, exposure to environmental or occupational toxins, or neurotoxic drug treatment . Common painful polyneuropathy included painful diabetes peripheral neuropathy (PDPN), chemotherapy induced peripheral neuropathy (CIPN), post radiotherapy pain, post-operative/traumatic pain, human immunodeficiency virus (HIV) infection painful polyneuropathy, etc. The prevalence of PDPN is 8.0%~26.0% , which is more likely to be chronic in type 2 diabetes. The prevalence of CIPN is about 40.0% . 13.0% to 50.0% of HIV patients may develop HIV infectious painful polyneuropathy .
(ii). Clinical Characteristics
PDPN most commonly affects the ends of the limbs, particularly the feet, and typically presents with persistent pain characterized by pricking, burning, or electric-shock-like sensations. The pain is often most prominent at rest and night.
CIPN is clinically characterized by sensory disturbances and neuropathic pain, including symmetric pain and numbness in the hands and feet, tingling, burning, or electric-shock-like sensations, which often worsen at night. Patients frequently experience muscle weakness, with advanced stages leading to motor impairment, such as muscle atrophy or coordination deficits.
Post radiotherapy pain refers to the pain caused by direct or delayed damage to nerves, bones, or soft tissues within the irradiation field during radiation therapy for primary tumors or tumor metastases. The most common form is chronic neuropathy caused by radiation.
Postoperative or posttraumatic pain refers to pain that occurs or intensifies due to surgery or tissue damage (including various injuries such as burns). The area of pain is often located within the surgical margin or tissue injury range, the corresponding nerve innervation area, and the skin segment area corresponding to surgery or injury of the deep body or visceral tissues.
Another painful polyneuropathy, such as HIV-infected painful polyneuropathy, typically manifests as abnormal sensory pain in the distribution area of stocking-glove, mainly affecting the feet and hands. Vitamin B deficiency causes painful polyneuropathy, with clinical manifestations mainly including lower limb burning pain or numbness, progressive aggravation, decreased muscle strength, and even muscle atrophy, seriously affecting walking. Patients with alcoholic polyneuropathy often complain of burning or numbness in the soles of the feet, fever, and spasmodic pain in the gastrocnemius muscle.
(iii). Diagnostic Criteria
PDPN is diagnosed according to the Toronto Consensus Criteria for Diabetic Neuropathy : Possible existence of PDPN History of diabetes, PDPN related symptoms, such as painful stocking-glove distribution sensory changes; Probable PDPN is diagnosed when two or more of the following criteria are met: neuropathic pain symptoms, distal sensory impairment, and/or diminished/absent ankle reflexes.; Definitive diagnosis of PDPN requires abnormal nerve conduction studies (NCS). If NCS findings are normal, small fiber nerve function testing may be employed to support diagnosis.
CIPN There are no clearly established diagnostic criteria. Chemotherapeutic agents, particularly oxaliplatin- or paclitaxel-based regimens, are more likely to induce neuropathic pain, which is more severe and lasts longer. It is recommended to use cotton swabs or wooden sticks to evaluate tactile sensation, use hot and cold objects to evaluate thermal sensation, and use tuning fork tests to evaluate vibration sensation.
Other painful polyneuropathy There are no clearly established diagnostic criteria, and diagnosis is mainly based on medical history and clinical characteristics. Vitamin B complex deficiency is common in obesity, tumors, and long-term chronic gastroenteritis after gastrointestinal surgery. Alcohol-related polyneuropathy is more common in chronic heavy alcohol use for 10 years or more.
4.1.4. Neuropathic Pain After Peripheral Nerve Injury
(i). Definition
NP after peripheral nerve injury is a persistent or recurrent NP caused by peripheral neuropathy . Peripheral nerve injury is often caused by trauma such as traction injury, compression injury, and cutting injury. Incomplete or poor functional recovery after nerve damage can lead to NP. Depending on the affected area, NP after peripheral nerve injury can be divided into phantom limb pain, residual limb pain, nerve entrapment syndrome, brachial plexus nerve injury, and other nerve trunk injuries . Residual limb pain refers to pain occurring in the remaining stump after amputation, whereas phantom limb pain is the perception of pain in the amputated portion of the limb. The incidence of NP following peripheral nerve injury ranges from 8.0% to 26.0% .
(ii). Clinical Characteristics
NP resulting from peripheral nerve injury is characterized by prolonged duration and complex clinical manifestations, including sensory deficits, motor dysfunction, and autonomic nervous system disturbances. The clinical manifestations mainly include spontaneous pain, allodynia, hyperalgesia, persistent pain, or paresthesia. The pain may manifest in various forms, including lancinating, lightning-like, burning, tearing, shooting, electric-shock-like, or needle-pricking sensations. These diverse presentations can be either deep-seated or superficial, with most patients experiencing two or more distinct pain qualities simultaneously. Symptom exacerbation commonly occurs with physical activity, fatigue, psychological stress, or changes in environmental/climatic conditions. Patients may experience symptoms such as muscle spasms, stiffness, weakness, and atrophy. Physical examination shows decreased muscle tone, muscle atrophy, weakened or absent tendon reflexes, and sensory abnormalities. Notably, the pain often persists chronically even after resolution of the original etiology, complete tissue healing, or effective disease control, significantly impairing quality of life and frequently accompanied by affective disorders.
(iii). Diagnosis
The diagnosis of NP after peripheral nerve injury primarily relies on a detailed understanding of the medical history, systematic physical examination, and necessary neurophysiological testing, as no unified diagnostic criteria currently exist. The main diagnostic standards currently employed are as follows : The medical history clearly indicates the presence of related trauma or diseases in the peripheral nervous system; The onset of pain shows a clear temporal correlation with the traumatic incident; The pain distribution corresponds to the anatomical territory of somatosensory nerves; The neurological examination confirms objective signs consistent with nerve damage or disease; At least one auxiliary examination confirms the presence of related damage or disease in the somatosensory system.
4.1.5. Painful Radiculopathy
(i). Definition
Painful radiculopathy is a disease that causes peripheral neuropathic pain through nerve root lesions, characterized by persistent or recurrent pain resulting from pathologies affecting cervical, thoracic, lumbar, or sacral nerve roots, and classified as peripheral neuropathic pain. Degenerative spinal changes are the most common cause of painful radiculopathy, while trauma, tumors, neoplastic meningitis, infections, hemorrhage or ischemia, diabetes, rheumatoid arthritis, and iatrogenic conditions may also contribute .
(ii). Clinical Characteristics
The main clinical manifestations are spontaneous pain, hyperalgesia, and sensory abnormalities in the corresponding nerve root innervation area. The pain may present as persistent or paroxysmal, with qualities including lancinating, burning, tearing, electric-shock-like, or needle-stabbing sensations, often leading to functional impairment.
(iii). Diagnosis
The diagnosis primarily relies on medical history, physical examination, assessment scales, imagings, neurophysiological tests, and diagnostic interventions.
4.1.6. Glossopharyngeal Neuralgia
(i). Definition
Glossopharyngeal neuralgia (GPN) is a disease characterized by brief, paroxysmal episodes of unilateral pain, occurring in the distribution area of the glossopharyngeal nerve (mandible, ears, tonsil fossa, and tongue base). When involving the pharyngeal and auricular branches of the vagus nerve, symptoms are often triggered by swallowing. The annual incidence rate of GPN is about 0.7/100000, with increasing prevalence in older populations.
(ii). Diagnosis
The diagnostic criteria for GPN in ICHD-3 are as follows.
Recurrent and paroxysmal pain attacks in the distribution area of unilateral glossopharyngeal nerve.
Pain should have all of the following characteristics: The pain lasts from a few seconds to 2 minutes; The degree of pain is severe; The pain is characterized as sharp, stabbing, or electric shock-like sensations; Pain is triggered or exacerbated by coughing, yawning, swallowing, or speaking.
Pain cannot be explained by any other ICHD-3 diagnosis.
(iii). Differential Diagnosis
The diagnosis requires excluding other diseases that may cause similar symptoms, such as TN, temporal arteritis, temporomandibular joint dysfunction, etc. Diagnostic blockade via pharyngeal topical anesthesia is commonly used in clinical practice to assist in differential diagnosis.
4.2. Central Neuropathic Pain
4.2.1. Central Neuropathic Pain Associated with Spinal Cord Injury
(i). Definition
The central NP associated with spinal cord injury, abbreviated as spinal cord injury pain (SCIP), is one of the common sequelae of spinal cord injury. SCIP is divided into two categories.
The first category is nociceptive pain, which can be further divided into musculoskeletal pain and visceral pain.
The second category is neuropathic pain, which can be further divided into pain above the injury plane, pain below the injury plane, and pain below the injury plane. Numerous studies have shown that approximately 2/3 of spinal cord injury patients develop SCIP. Apart from motor dysfunction and sphincter disturbances, SCIP is often the greatest source of suffering for these patients.
(ii). Clinical Characteristics
Musculoskeletal pain is the most common type of pain during the acute phase after spinal cord injury. Pain episodes are often associated with muscle contractions, limb movements, or changes in posture, and may radiate into the limbs and trunk.
Visceral pain following spinal cord injury is primarily characterized in the thoracic, abdominal, or pelvic regions. The pain is typically diffuse, poorly localized, and often described as dull, colicky, or aching. It usually emerges months or even years after the spinal cord injury and tends to occur intermittently.
Spinal cord injury induced NP is often characterized by severe, shooting, burning, cutting, or stabbing pain, frequently accompanied by band-like dysesthesia.
(iii). Diagnosis
Clear history of spinal cord injury, including traumatic spinal cord injury, iatrogenic spinal cord injury, or other confirmed causes.
At least one auxiliary examination confirming that the pain corresponds to a neuroanatomical distribution.
At least one auxiliary examination demonstrating relevant structural damage or pathology.
4.2.2. Central Neuropathic Pain Associated with Brain Injury
(i). Definition
Chronic pain is a common sequela and one of the most frequent complaints in patients with traumatic brain injury (TBI). The prevalence of chronic pain exceeds 50.0% in TBI patients, reaching as high as 75.0% in mild TBI cases . In most instances, chronic pain localizes to the bodily region of tissue injury. However, some chronic pain occurs in non-traumatized areas or body parts without apparent pathology, potentially arising from central mechanisms—termed ‘central pain’ . Post-TBI chronic pain encompasses headache, musculoskeletal pain, central pain, and other types .
(ii). Clinical Characteristics
Chronic headache following brain injury most commonly manifests in three types: tension-type headache, migraine, and mixed symptoms of both. The headaches are typically of moderate intensity, with 65.0% of patients describing bilateral pain localized to the frontal region, characterized by a combination of throbbing and pressure-like qualities. Among moderate to severe TBI patients, approximately 12.0% may develop complex regional pain syndrome (CRPS), primarily presenting with limb spasms. Some TBI patients experience delayed-onset pain syndromes, often emerging weeks to months post-injury, predominantly unilateral, with persistent pain and paroxysmal exacerbations.
(iii). Diagnosis
The diagnosis is established based on a history of TBI, combined with characteristic symptoms, physical examination findings, and relevant imaging studies.
4.2.3. Central Post Stroke Pain
(i). Overview
Central Post-Stroke Pain (CPSP) refers to somatic pain and sensory abnormalities associated with ischemic or hemorrhagic stroke lesions. The incidence of CPSP ranges from 1.0% to 12.0% , typically emerging 3–6 months post-stroke. The development of CPSP is closely linked to sensory deficits, with younger age, smoking, depression, and stroke severity identified as high-risk factors .
(ii). Clinical Characteristics
The pain is often located in the contralateral limb, face, or trunk of the central lesion, and may also occur in ipsilateral hemiparesis, but the pain symptoms are milder than those on the contralateral side. The pain manifests with qualities such as burning, stabbing, lightning-like, crushing, freezing, or tearing sensations. The location of pain is basically consistent with the distribution of sensory abnormalities.
(iii). Diagnosis
Diagnosis primarily relies on stroke history, pain characteristics, and sensory abnormality symptoms, while excluding other potential conditions. The diagnostic criteria for CPSP proposed by Klit et al. include the following essential components.
Pain localized to somatic areas corresponding to the central nervous system lesion.
History of stroke, with pain onset coinciding with or following the stroke.
Clinical examination reveals sensory deficits consistent with the lesion.
Neuroimaging confirms a relevant causative lesion.
Exclusion of alternative pain etiologies.
4.2.4. Central Neuropathic Pain Associated with Multiple Sclerosis
(i). Definition
Multiple sclerosis (MS) related central NP is a chronic inflammatory demyelinating disease of the central nervous system, frequently manifests with pain secondary to neurological damage. Pain is highly prevalent in MS, typically resulting from neurological damage, with the majority being central NP and a minority presenting as peripheral neuropathic pain or musculoskeletal pain. Between 50.0% and 75.0% of MS patients experience chronic pain at some point during the disease course, and in some cases, pain may be the initial symptom of MS. The type of pain in MS patients generally correlates with the specific regions of the nervous system affected.
(ii). Clinical Characteristics
Painful numbness is the most common type of pain in MS, present in 45.0% of patients. It is characterized by persistent burning pain that worsens with high temperatures or weather changes, and may be accompanied by allodynia and hyperalgesia. The pain is typically bilateral, affecting the legs and feet, and is more severe at night. Physical activity can exacerbate the pain. Patients with primary progressive MS may also experience brief, electric-shock-like or radiating pain lasting less than 2 seconds in the posterior neck, lower back, or other body areas, triggered by neck flexion and immediately relieved upon cessation of movement.
(iii). Diagnosis
The diagnosis of MS is primarily based on comprehensive analysis of clinical symptoms and ancillary test results, while requiring exclusion of other potential diseases.
5. Treatment
5.1. Treatment Principles
NP is a persistent condition that may involve recurrent exacerbations, requiring long-term therapeutic management. NP treatment should be based on safety, efficacy, and cost-effectiveness, guided by the principles of ‘moving the checkpoint forward, early intervention, prevention of sensitization, and prevention of chronic pain’. Pharmacotherapy is generally the first-line approach, with timely incorporation of minimally invasive procedures when indicated, and general treatment, physical therapy, traditional Chinese medicine treatment, psychological therapy, etc. should be combined as appropriate.
5.2. Treatment Methods
5.2.1. General Treatment
Environmental factors and emotional changes can worsen NP, highlighting the importance of patient education and self-management. Patients should maintain healthy habits, avoiding sleep deprivation, overexertion, excessive eating/drinking, smoking, and overly spicy foods. Guide patients to self-regulate their emotions. Regular exercise and social engagement are encouraged. A strong, trusting physician-patient relationship fosters better treatment adherence and outcomes. Evidence-based recommendations for general therapies, including quality grading and strength of evidence, are summarized in Table 2 .
Table 2. Evidence Quality and Recommendation Strength for General Therapies.

Intervention

Evidence Level

Recommendation Strength

Health education

A

1

Self-management

, 29]

A

1

5.2.2. Pharmacotherapy
Pharmacotherapy forms the foundation of treatment and should utilize recommended drugs at effective doses . Combination therapy is well-tolerated and can enhance analgesic efficacy in patients with inadequate pain control from monotherapy . Upon achieving effective pain relief, abrupt discontinuation should be avoided, with maintenance treatment continued for at least 2 weeks .
(i) Common drug classification
Calcium channel-targeting agents: Primary representative medications include pregabalin, gabapentin, crisugabalin, and mirogabalin .
Sodium channel modulators: Key representative drugs comprise carbamazepine, oxcarbazepine, lidocaine, and bulleyaconitine A.
Tricyclic antidepressants (TCAs): The main representative drugs include amitriptyline, imipramine, desipramine, and nortriptyline. However, there is little evidence to support the use of imipramine , desipramine , or nortriptyline for treating NP.
Serotonin and norepinephrine reuptake inhibitors (SNRIs): Key representative drugs include duloxetine and venlafaxine.
Opioids: Main representative drugs include morphine, oxycodone, fentanyl, buprenorphine, tapentadol, methadone, and hydromorphone. However, there is insufficient evidence to support or refute the efficacy of morphine , fentanyl , or hydromorphone in NP treatment.
NMDA receptor antagonists (e.g., ketamine, memantine, dextromethorphan).
Botulinum toxin type A (BTX-A) and the novel anti-inflammatory drug tetrandrine can be effectively used in the comprehensive management of NP.
Platelet-rich plasma (PRP) : Perineural PRP injection is an effective therapy for alleviating pain and numbness in PDPN and improving peripheral nerve function.
Other medications: such as tramadol, capsaicin patch, cannabinoids, antiepileptic drugs (e.g., lamotrigine, topiramate), vaccinia virus-inoculated rabbit skin inflammatory extract, traditional Chinese medicines (e.g., Jingshu Granules ), glucocorticoids, vitamins, muscle relaxants (e.g., baclofen), etc.
(ii) The quality grading and recommendation strength of evidence-based medicine for common NP treatment drugs (Table 3).
Pregabalin has the widest and highest level of evidence support. For PHN , PDPN , CIPN, post radiotherapy pain , postoperative or posttraumatic pain , SCIP , CPSP, the evidence level of pregabalin is A1. In contrast, the strength of evidence for Gabapentin varies. Its evidence level for TN is B2 , but its evidence level for PHN and PDPN is A1. For CIPN and post radiotherapy pain , the evidence level drops to B1. For SCIP, the level of evidence returned to A1, but for CPSP , the level of evidence was downgraded to B1. The new drug Crisugabalin showed A1 level evidence for both TN and PHN . Another new drug, Mirogabalin, has an evidence level of A1 for PHN and PDPN , B1 for CIPN, A1 for postoperative or posttraumatic pain and SCIP , and B1 for CPSP .
In the category of sodium channel blockers, carbamazepine , is a classic A1 level recommended drug for TN, but its evidence level for PHN is only B2 . Oxcarbazepine is also an A1 level recommended drug for TN , but the evidence level for PHN is B2 , and the evidence level for SCIP is also B2 . Lidocaine has an evidence level of B2 for TN but shows A1 evidence for PHN , and an evidence level of B2 for SCIP .
Among tricyclic antidepressants, amitriptyline has an evidence level of B2 for PHN , but an evidence level of A1 for SCIP . Duloxetine has extensive A1 level evidence among SNRIs, including PDPN , CIPN , postoperative or posttraumatic pain , SCIP, and CPSP . The evidence level of Venlafaxine for PDPN and CIPN is A1, but the evidence level for SCIP is B2 .
In opioid drugs, the evidence level for buprenorphine on PHN and PDPN is B2.In NMDA receptor antagonists, the evidence level of ketamine for PHN and SCIP[62, 63] is B2, with references and , respectively. In the category of others, the level of evidence for multiple drugs is generally not high. Type A botulinum toxin (BTX-A) has an evidence level of B2 in TN, PHN , PDPN , and SCIP . 8% capsaicin has an evidence level of B2 in PHN PDPN , CIPN , and SCIP . Lamotrigine has an evidence level of B2 in TN , SCIP , and CPSP . The evidence level of vaccinia virus immune globulin for PHN is A1 , but for PDPN is B2 .
(iii) Recommended NP drug therapy (Table 4 ).
(iv) TN drug therapy recommendations (Table 5 ).
(v) Recommendations for drug treatment of diabetes peripheral neuropathic pain (Table 6).
(vi) Recommendations for drug treatment of central neuropathic pain (Table 7).
Table 3. Evidence Quality Grading and Recommendation Strength for Pharmacotherapy.

Drug Class

Drug Name

TN

PHN

Painful Polyneuropathy

Central Neuropathic Pain (Spinal Cord Injury)

CPSP

PDPN

CIPN

Post radiotherapy pain

Postoperative or posttraumatic pain

Calcium Channel Modulators

Pregabalin

A1

A1

A1

A1

-54]

A1

A1

A1

Gabapentin

B2

A1

, 43, 73-75]

A1

B1

B1

A1

, 66]

B1

Crisugabalin

A1

A1

Mirogabalin

A1

, 84]

A1

B1

, 92]

A1

A1

, 94]

B1

Sodium Channel Blockers

Carbamazepine

A1

B2

Oxcarbazepine

A1

, 99, 101, 102]

B2

, 104]

B2

Lidocaine

B2

, 97]

A1

, 107]

B2

Tricyclic Antidepressants

Amitriptyline

B2

A1

, 66, 108, 109]

SNRIs

Duloxetine

A1

, 45, 47, 49, 50, 76, 104, 110]

A1

A1

A1

A1

Venlafaxine

A1

A1

B2

Opioids

Buprenorphine

B2

B2

NMDA Receptor Antagonists

Ketamine

B2

B2

, 63]

Others

BTX-A

B2

, 120-124]

B2

B2

, 130]

B2

, 63, 131, 132]

8% Capsaicin

B2

, 134]

B2

B2

B2

Lamotrigine

B2

B2

B2

Vaccinia Virus Immune Globulin

A1

B2

Abbreviations: TN, trigeminal neuralgia; PHN, postherpetic neuralgia; PDPN, painful diabetes peripheral neuropathy; CIPN, chemotherapy induced peripheral neuropathy; CRPS, complex regional pain syndrome; BTX-A, botulinum toxin type A.
Table 4. Recommended Pharmacotherapy for Neuropathic Pain.

Treatment Recommendation

Recommended Drugs

First-line Therapy

Calcium channel modulators

, Tricyclic antidepressants (tcas), Serotonin-norepinephrine reuptake inhibitors (snris) , Topical lidocaine

Second-line Therapy

Lidocaine patches

, High-concentration capsaicin patches , Tramadol , Botulinum toxin type A , Strong opioids

Third-line Therapy

Strong opioids

, Botulinum toxin type A , Tramadol , Cannabinoids

Fourth-line Therapy

Methadone

, Tapentadol , Topical lidocaine , Botulinum toxin

Table 5. Recommended Pharmacotherapy for Trigeminal Neuralgia.

Treatment Recommendation

Commonly Used Drugs

First-line Therapy

Carbamazepine

, Oxcarbazepine

Second-line Therapy

Lamotrigine

, Baclofen , Gabapentin , Pregabalin

Table 6. Recommended Pharmacotherapy for Painful Diabetic Peripheral Neuropathy.

Treatment Recommendation

Commonly Used Drugs

First-line Therapy

Duloxetine, Venlafaxine, Amitriptyline, Pregabalin, Gabapentin

Second-line Therapy

Tramadol, Morphine

Note: The National Medical Products Administration (NMPA) approved cligabalin and mirogabalin in 2024 for the treatment of painful diabetic peripheral neuropathy in adults.
Table 7. Recommended Pharmacotherapy for Central Neuropathic Pain.

Treatment Recommendation

Drugs

First-line Therapy

Gabapentin, Duloxetine, Tricyclic antidepressants (TCAs)

Second-line Therapy

Venlafaxine, Pregabalin, Lamotrigine, Tramadol

Third-line Therapy

Methadone, Oxycodone, Cannabinoids

5.2.3. Physical Therapy
Physical therapy is a common non-invasive, non-pharmacological treatment approach widely used in NP management. It primarily includes photobiomodulation therapy (PBMT), low-level laser therapy (LLLT), transcutaneous electrical nerve stimulation (TENS), scrambler therapy (ST), extracorporeal shockwave therapy (ESWT), repetitive transcranial magnetic stimulation (rTMS) , transcranial direct electrical stimulation (tDCS) , cryotherapy, whole-body vibration (WBV), yoga, etc. (Table 8 ).
Table 8. Evidence Quality Grading and Recommendation Strength for Physical Therapy Interventions.

Treatments

TN

PHN

Painful Polyneuropathy

Central Neuropathic Pain (Spinal Cord Injury)

CPSP

PDPN

CIPN

Postoperative or posttraumatic pain

Painful Radiculopathy

PBMT

B2

B2

B2

LLLT

B2

B2

B1

B2

TENS

A1

, 168]

A2

A1

, 172]

A2

, 174]

B2

A2

A2

ST

B2

ESWT

B2

B2

rTMS

B2

B2

, 186]

B2

A2

B2

A1

A2

tDCS

B2

B2

, 192]

A1

, 207]

B2

, 203]

Cryotherapy

B2

WBV

B2

, 211]

B2

Yoga

B2

B2

Abbreviations: PHN, Postherpetic neuralgia; PDPN, Painful diabetic peripheral neuropathy; CIPN: Chemotherapy-induced peripheral neuropathy; CPSP, Central post-stroke pain; PBMT, Photobiomodulation therapy; LLLT, Low-level laser therapy; TENS, Transcutaneous electrical nerve stimulation; ST, Scrambler therapy; ESWT, Extracorporeal shock wave therapy; rTMS, Repetitive transcranial magnetic stimulation; tDCS: Transcranial direct current stimulation; BWV, Whole-body vibration
5.2.4. Minimally Invasive Interventional Therapy
Minimally invasive intervention therapy refers to the use of imaging equipment such as CT, ultrasound, digital subtraction angiography (DSA), C-arm, etc. to guide the observation, diagnosis, and treatment of lesions in the body with the smallest incision path and minimal tissue damage. It mainly includes nerve block, nerve damage (chemical and physical damage), nerve regulation, etc. Specific treatment methods include stellate ganglion block (SGB), continuous radio frequency (CRF), pulsed radio frequency (PRF), pulsed combined continuous radio frequency (PCRF), and percutaneous balloon compression (PCF). Balloon compression (PBC), spinal cord electrical stimulation (SCS) . Deep brain stimulation (DBS) , motor cortex stimulation (MCS), peripheral nerve stimulation (PNS), dorsal root ganglion stimulation (DRGS), intrathecal drug delivery system (IDDS), etc. (Table 9).
Neuroblock (NB) has shown A1 levels of evidence for PHN and painful radiculopathy. Percutaneous balloon compression (PBC) is an A1 level recommended method for treating TN . Condensed radiofrequency (CRF) is also an A1 level method for treating TN . In addition, its evidence level for postoperative or post-traumatic pain is A2 , and its evidence level for painful radiculopathy is B2. Pulse radiofrequency (PRF) therapy has a wide range of A1 level evidence and is suitable for trigeminal TN, PHN , postoperative or posttraumatic pain , and painful radiculopathy . Pulse combined with condensing radiofrequency (PCRF) has also received A1 level recommendation in the treatment of TN [233, 238, 248-250]. Spinal cord electrical stimulation (SCS) has shown strong evidence support in multiple fields, with A1 level recommendations for PHN, PDPN , CIPN [261], postoperative or post-traumatic pain . However, for SCIP [266], the evidence level is weak, at B2 level. The deep brain stimulation (DBS) has B2 level evidence for postoperative or post-traumatic pain and SCIP , but with evidence level A1 for CPSP. The evidence strength for motor cortex stimulation (MCS) is generally not high, with a grade of B2 for TN and postoperative or post-traumatic pain , with A2 grade for CPSP. Peripheral nerve stimulation (PNS) has B2 level evidence for TN, and CIPN . But its evidence level for PHN and postoperative or post-traumatic pain is A1. The level of evidence for root ganglion stimulation (DRGS) is relatively low, with B1 grade for PDPNand B2 grade for CIPN and CPSP . The intrathecal drug infusion system (IDDS) only showed B2 level evidence in SCIP .
Table 9. Evidence Quality and Recommendation for Minimally Invasive Interventional Therapies.

Treatments

TN

PHN

Painful Polyneuropathy

Central Neuropathic Pain (Spinal Cord Injury)

CPSP

PDPN

CIPN

Postoperative or posttraumatic pain

Painful Radiculopathy

NB

A1

A1

, 224]

PBC

A1

CRF

A1

, 234]

A2

, 236]

B2

PRF

A1

, 238]

A1

A1

A1

PCRF

A1

248-250]

SCS

A1

A1

A1

A1

B2

DBS

B2

B2

A1

, 268]

MCS

B2

B2

A2

PNS

B2

, 273]

A1

B2

A1

DRGS

B1

B2

B2

IDDS

B2

Abbreviations: PHN, Postherpetic neuralgia; PDPN, Painful diabetic peripheral neuropathy; CIPN, Chemotherapy-induced peripheral neuropathy; CPSP, Central post-stroke pain; PRC, Percutaneous balloon compression; CRF, Continuous radiofrequency; PRF, Pulsed radiofrequency; PCRF, Pulsed combined with continuous radiofrequency; SCS, Spinal cord stimulation; DBS, Deep brain stimulation; MCS, Motor cortex stimulation; PNS, Peripheral nerve stimulation; DRGS, Dorsal root ganglion stimulation; IDDS, Intrathecal drug delivery system.
5.2.5. Surgical Operation
Surgical treatments for NP primarily include nerve decompression and neurolysis (Table 10 ).
Table 10. Evidence Quality and Recommendation for Surgical Procedures.

Disease/Disorder

Treatment Method

Evidence Level

Recommendation Strength

Trigeminal neuralgia

MVD

A

1

SRS

A

1

Glossopharyngeal neuralgia

MVD

B

2

SRS

A

2

PDPN

Peripheral nerve decompression

A

1

Abbreviations: MVD, Microvascular decompression of cranial nerves; SRS, Stereotactic radiosurgery; PDPN, Painful diabetic peripheral neuropathy
(i) Nerve decompression primarily includes microvascular decompression (MVD) of cranial nerves and peripheral nerve decompression.
(ii) Neurolysis primarily includes stereotactic radiosurgery (SRS), punctate midline myelotomy (PMM), dorsal root entry zone (DREZ) lesioning, and peripheral neurectomy, among others.
5.2.6. Traditional Chinese Medicine (TCM) Therapy
Table 11. Evidence Quality and Recommendation for Traditional Chinese Medicine Therapies.

Treatments

TN

PHN

Painful Polyneuropathy

Central Neuropathic Pain (Spinal Cord Injury)

CPSP

PDPN

CIPN

Painful Radiculopathy

Acupuncture

A1

A1

A1

A1

A1

A2

A2

Warm Needling (Wen Zhen Jiu)

A1

, 319]

Heat-sensitive Moxibustion

B2

A2

, 322]

Electroacupuncture

B2

, 324]

B1

B2

, 327]

B2

B2

Fire Needling (Huo Zhen)

B2

Floating Acupuncture (Fu Zhen)

B2

, 333]

Plum-blossom Needle

B2

Acupotomy (Zhen Dao)

B2

B2

Moxibustion

B2

Cupping

B2

Acupoint Injection

B2

, 340]

Acupoint Sticking Therapy

B2

Jingshu Keli (Neck-soothing Granules)

B1

Puerarin Injection

B2

, 343]

Huangqi Guizhi Wuwu Tang (Astragalus and Cinnamon Twig Five-Substance Decoction)

B2

, 345]

Xuefu Zhuyu Tang (Blood Stasis-Expelling Decoction)

B2

Buyang Huanwu Tang (Decoction for Invigorating Yang)

B2

Shentong Zhuyu TangTangkuei and Myrrha

B2

Shaoyao Gancao Tang (made of P. lactiflora and G. uralensis)

B2

Abbreviations: PHN, Postherpetic neuralgia; PDPN, Painful diabetic peripheral neuropathy; CIPN, Chemotherapy-induced peripheral neuropathy; CPSP, Central post-stroke pain
Based on the theory of syndrome differentiation and treatment, TCM approaches for NP mainly include external therapies and internal therapies. External therapies primarily consist of acupuncture, electroacupuncture, fire needling, floating needling, plum-blossom needling, acupotomy, moxibustion, manual therapies (tuina, massage, guasha, cupping, etc.), and acupoint injection. Internal therapies mainly involve Chinese patent medicines and herbal preparations (Table 11 ).
5.2.7. Psychotherapy
Psychotherapy refers to a therapeutic process conducted by professionally trained physicians who apply specialized theories and techniques, based on establishing a strong physician-patient relationship. NP patients often have mental and psychological problems, so psychotherapy is one of the important treatment methods for NP, which can alleviate patients' psychological pressure and pain. The commonly used psychological therapy methods include cognitive-behavioral therapy (CBT) , mindfulness meditation (MM) , mindfulness based cognitive therapy (MBCT), mindfulness-based stress reduction (MBSR), acceptance and commitment therapy (ACT) , hypnosis , aromatherapy, etc. (Table 12 ). Psychological therapy can also be combined with other treatments to improve treatment effectiveness.
Table 12. Evidence Quality and Recommendation for Psychological Therapies.

Treatments

PHN

Painful Polyneuropathy

PDPN

Post radiotherapy pain

Postoperative pain

CBT

A1

MM

B2

A1

MBCT

A1

A1

MBSR

B2

Hypnotherapy

B2

Aromatherapy

B2

, 364]

Abbreviations: PHN, Postherpetic neuralgia; PDPN, Painful diabetic peripheral neuropathy; CBT, Cognitive behavioral therapy; MM, Mindfulness meditation; MBCT, Mindfulness-based cognitive therapy; MBSR, Mindfulness-based stress reduction.
Abbreviations

BTX-A

Botulinum Toxin Type A

BWV

Whole-Body Vibration

CBT

Cognitive Behavioral Therapy

CIPN

Chemotherapy Induced Peripheral Neuropathy

CNKI

China National Knowledge Infrastructure

CPSP

Central Post-Stroke Pain

CRF

Continuous Radiofrequency

CRPS

Complex Regional Pain Syndrome

DBS

Deep Brain Stimulation

DREZ

Dorsal Root Entry Zone

DRGS

Dorsal Root Ganglion Stimulation

ESWT

Extracorporeal Shock Wave Therapy

GRADE

Grading of Recommendations Assessment, Development and Evaluation

GPN

Glossopharyngeal Neuralgia

HIV

Human Immunodeficiency Virus

ICHD-3

The Third Edition of International Classification of Headache Disorders

IDDS

Intrathecal Drug Delivery System

LLLT

Low-Level Laser Therapy

LTP

Long Term Potentiation

MBCT

Mindfulness-Based Cognitive Therapy

MBSR

Mindfulness-Based Stress Reduction

MCS

Motor Cortex Stimulation

MM

Mindfulness Meditation

MS

Multiple Sclerosis

MVD

Microvascular Decompression of Cranial Nerves

NCS

Nerve Conduction Studies

NP

Neuropathic Pain

PBMT

Photobiomodulation Therapy

PCRF

Pulsed Combined with Continuous Radiofrequency

PDPN

Painful Diabetes Peripheral Neuropathy

PHN

Postherpetic Neuralgia

PMM

Punctate Midline Myelotomy

PNS

Peripheral Nerve Stimulation

PRC

Percutaneous Balloon Compression

PRF

Pulsed Radiofrequency

PRP

Platelet-Rich Plasma

RCTs

Randomized Controlled Trials

rTMS

repetitive Transcranial Magnetic Stimulation

SCIP

Spinal Cord Injury Pain

SCS

Spinal Cord Stimulation

SNRIs

Serotonin and Norepinephrine Reuptake Inhibitors

SRS

Stereotactic Radiosurgery

ST

Scrambler Therapy

TBI

Traumatic Brain Injury

TCAs

Tricyclic Antidepressants

tDCS

Transcranial Direct Current Stimulation

TENS

Transcutaneous Electrical Nerve Stimulation

TN

Trigeminal Neuralgia

Acknowledgments
Special thanks to Kan Houming from Macau University of Science and Technology for professional translation assistance.
Conflicts of Interest
All authors declare no conflicts of interest.
References
[1] Thouaye M, Yalcin I. Neuropathic pain: From actual pharmacological treatments to new therapeutic horizons [J] Pharmacol Ther, 2023, 251: 108546.
[2] Guyatt G, Oxman AD, Akl EA, et al. GRADE guidelines: 1. introduction GRADE evidence profiles and summary of findings tables [J]. J Clin Epidemiol, 2011, 64(4): 383394.
[3] Balshem H, Helfand M, Schünemann HJ, et al. GRADE guidelines: 3. rating the quality of evidence [J]. J Clin Epidemiol, 2011, 64(4): 401406.
[4] Jaeschke R, Guyatt GH, Dellinger P, et al. Use of GRADE grid to reach decisions on clinical practice guidelines when consensus is elusive [J]. BMJ, 2008, 337: a744.
[5] Kuner R, Flor H. Structural plasticity and reorganisation in chronic pain [J]. Nat Rev Neurosci, 2016, 18(1): 20-30.
[6] Moldovan M, Alvarez S, Romer Rosberg M, et al. Axonal voltage-gated ion channels as pharmacological targets for pain [J]. Eur J Pharmacol, 2013, 708(1-3): 105-112.
[7] Liu Y, Zhou LJ, Wang J, et al. TNF-α differentially regulates synaptic plasticity in the hippocampus and spinal cord by microglia-dependent mechanisms after peripheral nerve injury [J]. J Neurosci, 2017, 37(4): 871-881.
[8] Ossipov MH, Morimura K, Porreca F. Descending pain modulation and chronification of pain [J]. Curr Opin Support Palliat Care, 2014, 8(2): 143-151.
[9] Ong WY, Stohler CS, Herr DR. Role of the prefrontal cortex in pain processing [J]. Mol Neurobiol, 2019, 56(2): 1137-1166.
[10] Liu XG. Normalization of neuroinflammation: a new strategy for treatment of persistent pain and memory/emotional deficits in chronic pain [J]. J Inflamm Res, 2022, 15: 5201-5233.
[11] Functional Neurosurgery Group of Chinese Neurosurgical Society, Chinese Medical Association Functional Neurosurgery Expert Committee of Chinese Association of Neurosurgeons. Chinese expert consensus on diagnosis and treatment of trigeminal neuralgia [J]. Chinese Journal of Surgery, 2015, 53(9): 657-664.
[12] Expert Panel on PHN Diagnosis and Treatment Consensus. Chinese Expert Consensus on the Diagnosis and Treatment of Postherpetic Neuralgia [J]. Chinese Journal of Pain Medicine, 2016, 22(3): 161-167.
[13] Scholz J, Finnerup NB, Attal N, et al. Classification Committee of the Neuropathic Pain Special Interest Group (NeuPSIG). The IASP classification of chronic pain for ICD-11: chronic neuropathic pain [J]. Pain, 2019, 160(1): 53-59.
[14] Zuidema X, de Galan B, Brouwer B, et al. 4. Painful diabetic polyneuropathy [J]. Pain Pract, 2024, 24(2): 308-320.
[15] Bennett MI, Rayment C, Hjermstad M, et al. Prevalence and review [J]. Pain, 2012, 153(2): 359-365.
[16] Slawek DE. People living with HIV and the emerging field of chronic pain-what is known about epidemiology, etiology, and management [J]. Curr HIV/AIDS Rep, 2021, 18(5): 436-442.
[17] Franceschi R, Mozzillo E, Di Candia F, et al. A systematic review of the prevalence, risk factors and screening tools for autonomic and diabetic peripheral neuropathy in children, adolescents and young adults with type 1 diabetes [J]. Acta Diabetol, 2022, 59(3): 293-308.
[18] Jensen TS, Karlsson P, Gylfadottir SS, et al. Painful and non-painful diabetic neuropathy, diagnostic challenges and implications for future management [J]. Brain, 2021, 144(6): 1632-1645.
[19] Pain Diagnosis and Treatment Special Capacity Improvement Project Expert Group, National Health Commission Capacity Building and Continuing Education Center. Chinese Clinical Practice Guideline for Chronic Post-Traumatic Pain (2023 Edition) [J]. Chinese Journal of Pain Medicine, 2023, 19(4): 536-545.
[20] Chinese Expert Consensus on the Diagnosis and Treatment of Peripheral Neuropathic Pain [J]. Chinese Journal of Pain Medicine, 2020, 26(5): 321-328.
[21] Finnerup NB, Haroutounian S, Kamerman P, et al. Neuropathic pain: an updated grading system for research and clinical practice [J]. Pain, 2016, 157(8): 1599-1606.
[22] Kim JH, Ahn SH, Cho YW, et al. The relation between injury of the spinothalamocortical tract and central pain in chronic patients with mild traumatic brain injury [J]. J Head Trauma Rehabil, 2015, 30(6): E40-E46.
[23] Sodders MD, Martin AM, Coker J, et al. Acupuncture use for pain after traumatic brain injury: a NIDILRR Traumatic Brain Injury Model Systems cohort study [J]. Brain Inj, 2023, 37(6): 494-502.
[24] Kong KH, Woon VC, Yang SY. Prevalence of chronic pain and its impact on health-related quality of life in stroke survivors [J]. Arch Phys Med Rehabil, 2004, 85(1): 35-40.
[25] Smith JH, Bottemiller KL, Flemming KD, et al. Inability to self-report pain after a stroke: a population-based study [J]. Pain, 2013, 154(8): 1281-6.
[26] Klit H, Finnerup NB, Andersen G, et al. Central poststroke pain: a population-based study [J]. Pain, 2011, 152(4): 818-824.
[27] Ghavami H, Radfar M, Soheily S, et al. Effect of lifestyle interventions on diabetic peripheral neuropathy in patients with type 2 diabetes, result of a randomized clinical trial [J]. Agri, 2018, 30(4): 165-170.
[28] Egan KE, Caldwell GM, Eckmann MS. HIV neuropathy-a review of mechanisms, diagnosis, and treatment of pain [J]. Curr Pain Headache Rep, 2021, 25(8): 55.
[29] Reyhanıoglu DA, Yıldırım G, Sengun IŞ, et al. Effects of computer-based balance exercises on balance, pain, clinical presentation and nerve function in patients with diabetic peripheral neuropathy: a randomized controlled study [J]. J Musculoskelet Neuronal Interact, 2024, 24(2): 168-177. PMID: 38825999.
[30] Tesfaye S, Sloan G, Petrie J, et al. OPTION-DM trial group. Comparison of amitriptyline supplemented with pregabalin, pregabalin supplemented with amitriptyline, and duloxetine supplemented with pregabalin for the treatment of diabetic peripheral neuropathic pain (OPTION-DM): a multicentre, double-blind, randomised crossover trial [J]. Lancet, 2022, 400(10353): 680-690.
[31] Hearn L, Derry S, Phillips T, et al. Imipramine for neuropathic pain in adults [J]. Cochrane Database Syst Rev, 2014, 2014(5): CD010769.
[32] Hearn L, Moore RA, Derry S, et al. Desipramine for neuropathic pain in adults [J]. Cochrane Database Syst Rev, 2014, 2014(9): CD011003.
[33] Derry S, Wiffen PJ, Aldington D, et al. Nortriptyline for neuropathic pain in adults [J]. Cochrane Database Syst Rev, 2015, 1(1): CD011209.
[34] Cooper TE, Chen J, Wiffen PJ, et al. Morphine for chronic neuropathic pain in adults [J]. Cochrane Database Syst Rev, 2017, 5(5): CD011669.
[35] Derry S, Stannard C, Cole P, et al. Fentanyl for neuropathic pain in adults [J]. Cochrane Database Syst Rev, 2016, 10(10): CD011605.
[36] Stannard C, Gaskell H, Derry S, et al. Hydromorphone for neuropathic pain in adults [J]. Cochrane Database Syst Rev, 2016, 2016(5): CD011604.
[37] Duan XF, Yan F, He J. Clinical efficacy of tetrandrine tablets in patients with sciatica [J]. Chinese Journal of Clinical Healthcare, 2013, 16(5): 518-519.
[38] Hassanien M, Elawamy A, Kamel EZ, et al. Perineural platelet-rich plasma for diabetic neuropathic pain, could it make a difference? [J]. Pain Med, 2020, 21(4): 757-765.
[39] Hu J, Chen F, Qiu G, et al. Jingshu Keli for treating cervical spondylotic radiculopathy: the first multicenter, randomized, controlled clinical trial [J]. J Orthop Translat, 2020, 27: 44-56.
[40] Derry S, Bell RF, Straube S, et al. Pregabalin for neuropathic pain in adults [J]. Cochrane Database Syst Rev, 2019, 1(1): CD007076.
[41] Liu Q, Chen H, Xi L, et al. A randomized, double-blind, placebo-controlled trial to evaluate the efficacy and safety of pregabalin for postherpetic neuralgia in a population of Chinese patients [J]. Pain Pract, 2017, 17(1): 62-69.
[42] Gong R, Zhu SS, Zhu Y. Meta-analysis of pregabalin versus gabapentin for postherpetic neuralgia [J]. Practical Pharmacy and Clinical Remedies, 2023, 26(5): 390-400.
[43] Ding Y, Xie H, Ge WH. Meta-analysis comparing the efficacy and safety of pregabalin versus gabapentin for postherpetic neuralgia [J]. Pharmaceutical and Clinical Research, 2019, 27(1): 57-60.
[44] Liampas A, Rekatsina M, Vadalouca A, et al. Pharmacological management of painful peripheral neuropathies: a systematic review [J]. Pain Ther, 2021, 10(1): 55-68.
[45] Irving G, Tanenberg RJ, Raskin J, et al. Comparative safety and tolerability of duloxetine vs. pregabalin vs. duloxetine plus gabapentin in patients with diabetic peripheral neuropathic pain [J]. Int J Clin Pract, 2014, 68(9): 1130-1140.
[46] Neuropathy Study Group of Chinese Diabetes Society, Primary Diabetes Prevention and Management Office of National Basic Public Health Service Project. National guidelines for diagnosis and treatment of primary diabetic neuropathy (2024 edition) [J]. Chinese Journal of Diabetes Mellitus, 2024, 16(5): 496 511.
[47] Jingxuan L, Litian M, Jianfang F. Different drugs for the treatment of painful diabetic peripheral neuropathy: a meta-analysis [J]. Front Neurol, 2021, 12: 682244.
[48] Parsons B, Li C. The efficacy of pregabalin in patients with moderate and severe pain due to diabetic peripheral neuropathy [J]. Curr Med Res Opin, 2016, 32(5): 929-937.
[49] D'Souza RS, Barman R, Joseph A, et al. Evidence-based treatment of painful diabetic neuropathy: a systematic review [J]. Curr Pain Headache Rep, 2022, 26(8): 583-594.
[50] Han JP, Tian RX, Fan BF, et al. Meta-analysis comparing the efficacy and safety of duloxetine versus pregabalin for diabetic peripheral neuropathic pain [J]. Chinese Journal of Pain Medicine, 2020, 26(5): 357-361.
[51] Salehifar E, Janbabaei G, Hendouei N, et al. Comparison of the efficacy and safety of pregabalin and duloxetine in taxane induced sensory neuropathy: a randomized controlled trial [J]. Clin Drug Investig, 2020, 40(3): 249-257.
[52] Kouri M, Rekatsina M, Vadalouca A, et al. Pharmacological management of neuropathic pain after radiotherapy in head and neck cancer patients: a systematic review [J]. J Clin Med, 2022, 11(16): 4877.
[53] Jiang J, Li Y, Shen Q, et al. Effect of pregabalin on radiotherapy-related neuropathic pain in patients with head and neck cancer: a randomized controlled trial [J]. J Clin Oncol, 2019, 37(2): 135-143.
[54] Lefebvre T, Tack L, Lycke M, et al. Effectiveness of adjunctive analgesics in head and neck cancer patients receiving curative (chemo-) radiotherapy: a systematic review [J]. Pain Med, 2021, 22(1): 152-164.
[55] Maleki MS, Zamani Z, Amiri R, et al. Pregabalin in patients with post-traumatic peripheral neuropathic pain: a meta-analysis of randomized controlled trials [J]. Pain Pract, 2023, 23(6): 595-602.
[56] Mishriky BM, Waldron NH, Habib AS. Impact of pregabalin on acute and persistent postoperative pain: a systematic review and meta-analysis [J]. Br J Anaesth, 2015, 114(1): 10-31.
[57] Zhang L, Zhang H. The efficacy of pregabalin for pain control after thoracic surgery: a meta-analysis [J]. J Cardiothorac Surg, 2024, 19(1): 4.
[58] Matsutani N, Dejima H, Takahashi Y, et al. Pregabalin reduces post-surgical pain after thoracotomy: a prospective, randomized, controlled trial [J]. Surg Today, 2015, 45(11): 1411-1416.
[59] Yu X, Liu T, Zhao D, et al. Efficacy and safety of pregabalin in neuropathic pain followed spinal cord injury: a review and meta-analysis of randomized controlled trials [J]. Clin J Pain, 2019, 35(3): 272-278.
[60] Davari M, Amani B, Amani B, et al. Pregabalin and gabapentin in neuropathic pain management after spinal cord injury: a systematic review and meta-analysis [J]. Korean J Pain, 2020, 33(1): 3-12.
[61] Allison DJ, Ahrens J, Mirkowski M, et al. The effect of neuropathic pain treatments on pain interference following spinal cord injury: a systematic review [J]. J Spinal Cord Med, 2024, 47(4): 465-476.
[62] Ling HQ, Chen ZH, He L, et al. Comparative efficacy and safety of 11 drugs as therapies for adults with neuropathic pain after spinal cord injury: a bayesian network analysis based on 20 randomized controlled trials [J]. Front Neurol, 2022, 13: 818522.
[63] Koukoulithras I, Alkhazi A, Gkampenis A, et al. A systematic review of the interventions for management of pain in patients after spinal cord injury [J]. Cureus, 2023, 15(7): E42657.
[64] Canavan C, Inoue T, McMahon S, et al. The efficacy, adverse events, and withdrawal rates of the pharmacological management of chronic spinal cord injury pain: a systematic review and meta-analysis [J]. Pain Med, 2022, 23(2): 375-395.
[65] Loh E, Guy SD, Mehta S, et al. The CanPain SCI clinical practice guidelines for rehabilitation management of neuropathic pain after spinal cord: introduction, methodology and recommendation overview [J]. Spinal Cord, 2016, 54(Suppl 1): S1-S6.
[66] Loh E, Mirkowski M, Agudelo AR, et al. The CanPain SCI clinical practice guidelines for rehabilitation management of neuropathic pain after spinal cord injury: 2021 update [J]. Spinal Cord, 2022, 60(6): 548-566.
[67] Xu H, Guan M, Chen Y, et al. Efficacy and safety of pregabalin vs carbamazepine in patients with central post-stroke pain [J]. Neurol Res, 2024, 46(3): 291-296.
[68] Bo Z, Jian Y, Yan L, et al. Pharmacotherapies for central post-stroke pain: a systematic review and network meta-analysis [J]. Oxid Med Cell Longev, 2022, 2022: 3511385.
[69] Chen KY, Li RY. Efficacy and safety of different antidepressants and anticonvulsants in central poststroke pain: a network meta-analysis and systematic review [J]. PLoS One, 2022, 17(10): e0276012.
[70] Zhao X, Ge S. The efficacy and safety of gabapentin vs. carbamazepine in patients with primary trigeminal neuralgia: a systematic review and meta-analysis [J]. Front Neurol, 2023, 14: 1045640.
[71] Cui HZ, Duan HB, Yang L, et al. Meta-analysis of efficacy and safety of gabapentin versus carbamazepine for trigeminal neuralgia [J]. Chinese Journal of Clinical Research, 2019, 32(12): 1639-1645.
[72] Yuan M, Xu LJ, Xiao ZL, et al. Systematic review of efficacy and safety of gabapentin versus carbamazepine for trigeminal neuralgia [J]. China Pharmacy, 2014, 25(40): 3795-3799.
[73] Wiffen PJ, Derry S, Bell RF, et al. Gabapentin for chronic neuropathic pain in adults [J]. Cochrane Database Syst Rev, 2017, 6(6): CD007938.
[74] Wang J, Zhu Y. Different doses of gabapentin formulations for postherpetic neuralgia: a systematical review and meta-analysis of randomized controlled trials [J]. J Dermatolog Treat, 2017, 28(1): 65-77.
[75] Zhang M, Gao CX, Ma KT, et al. A meta-analysis of therapeutic efficacy and safety of gabapentin in the treatment of postherpetic neuralgia from randomized controlled trials [J]. Biomed Res Int, 2018, 2018: 7474207.
[76] Jiang L, Xiong Y, Cui J. Comparison of the efficacy and safety of duloxetine and gabapentin in diabetic peripheral neuropathic pain: a meta-analysis [J]. Contrast Media Mol Imaging, 2022, 2022: 4084420.
[77] Asrar MM, Kumari S, Sekhar BC, et al. Relative efficacy and safety of pharmacotherapeutic interventions for diabetic peripheral neuropathy: a systematic review and bayesian network meta analysis [J]. Pain Physician, 2021, 24(1): E1-E14. PMID: 33400429.
[78] Ko YC, Lee CH, Wu CS, et al. Comparison of efficacy and safety of gabapentin and duloxetine in painful diabetic peripheral neuropathy: a systematic review and meta-analysis of randomised controlled trials [J]. Int J Clin Pract, 2021, 75(11): e14576.
[79] Zhang XL, Yuan HY, Zhang XH. Meta-analysis of gabapentin for diabetic peripheral neuropathic pain [J]. Drug Evaluation Research, 2015, 38(5): 557-562.
[80] Aghili M, Zare M, Mousavi N, et al. Efficacy of gabapentin for the prevention of paclitaxel induced peripheral neuropathy: a randomized placebo controlled clinical trial [J]. Breast J, 2019, 25(2): 226-231.
[81] Zhang DY, Lei TC, Qin LY, et al. Efficacy and safety of crisugabalin (HSK16149) in adults with postherpetic neuralgia: a phase 3 randomized clinical trial [J]. JAMA Dermatol, Published online September 25, 2024.
[82] Guo X, Zhang T, Yuan G, et al. GABA analogue hsk16149 in Chinese patients with diabetic peripheral neuropathic pain: a phase 3 randomized clinical trial [J]. JAMA Netw Open, 2024, 7(8): e2425614.
[83] Kato J, Matsui N, Kakehi Y, et al. Mirogabalin for the management of postherpetic neuralgia: a randomized, double-blind, placebo controlled phase 3 study in Asian patients [J]. Pain, 2019, 160(5): 1175-1185.
[84] Kato J, Matsui N, Kakehi Y, et al. Long-term safety and efficacy of mirogabalin in Asian patients with postherpetic neuralgia: results from an open-label extension of a multicenter randomized, double-blind, placebo-controlled trial [J]. Medicine (Baltimore), 2020, 99(36): e21976.
[85] Alyoubi RA, Alshareef AA, Aldughaither SM, et al. Efficacy and safety of mirogabalin treatment in patients with diabetic peripheral neuropathic pain: a systematic review and meta-analysis of randomised controlled trials [J]. Int J Clin Pract, 2021, 75(5): e13744.
[86] Merante D, Rosenstock J, Sharma U, et al. DS-5565-A-U201 US Phase 2 study investigators. efficacy of mirogabalin (DS-5565) on patient-reported pain and sleep interference in patients with diabetic neuropathic pain: secondary outcomes of a Phase II Proof-of-Concept study [J]. Pain Med, 2017, 18(11): 2198-2207.
[87] Vinik A, Rosenstock J, Sharma U, et al. DS5565-A-U201 US Phase II study investigators. efficacy and safety of mirogabalin (DS-5565) for the treatment of diabetic peripheral neuropathic pain: a randomized, double-blind, placebo- and active comparator controlled, adaptive proof-of-concept phase 2 study [J]. Diabetes Care, 2014, 37(12): 3253-3261.
[88] Baba M, Matsui N, Kuroha M, et al. Mirogabalin for the treatment of diabetic peripheral neuropathic pain: a randomized, double-blind, placebo-controlled phase III study in Asian patients [J]. J Diabetes Investig, 2019, 10(5): 1299-1306.
[89] Kato J, Baba M, Kuroha M, et al. Safety and efficacy of mirogabalin for peripheral neuropathic pain: pooled analysis of two pivotal phase iii studies [J]. Clin Ther, 2021, 43(5): 822-835, E16.
[90] Baba M, Matsui N, Kuroha M, et al. Long-term safety and efficacy of mirogabalin in Asian patients with diabetic peripheral neuropathic pain [J]. J Diabetes Investig, 2020, 11(3): 693-698.
[91] Sugimoto M, Takagi T, Suzuki R, et al. Mirogabalin vs pregabalin for chemotherapy-induced peripheral neuropathy in pancreatic cancer patients [J]. BMC Cancer, 2021, 21(1): 1319.
[92] Misawa S, Denda T, Kodama S, et al. MiroCIP study group. Efficacy and safety of mirogabalin for chemotherapy-induced peripheral neuropathy: a prospective single-arm trial (MiroCIP study) [J]. BMC Cancer, 2023, 23(1): 1098.
[93] Ushida T, Katayama Y, Hiasa Y, et al. Mirogabalin for central neuropathic pain after spinal cord injury: a randomized, double-blind, placebo-controlled, phase 3 study in asia [J]. Neurology, 2023, 100(11): E1193-E1206.
[94] Ushida T, Katayama Y, Hiasa Y, et al. Long-term safety and efficacy of mirogabalin for central neuropathic pain: a multinational, phase 3, 52-week, open-label study in Asia [J]. Pain Ther, 2023, 12(4): 963-978.
[95] Rana MH, Khan AAG, Khalid I, et al. Therapeutic approach for trigeminal neuralgia: a systematic review [J]. Biomedicines, 2023, 11(10): 2606.
[96] Yang F, Lin Q, Dong L, et al. Efficacy of 8 different drug treatments for patients with trigeminal neuralgia: a network meta-analysis [J]. Clin J Pain, 2018, 34(7): 685-690.
[97] Peterson-Houle GM, AbdelFattah MR, Padilla M, et al. Efficacy of medications in adult patients with trigeminal neuralgia compared to placebo intervention: a systematic review with meta-analyses [J]. J Dent Anesth Pain Med, 2021, 21(5): 379-396.
[98] Chong MS, Bahra A, Zakrzewska JM. Guidelines for the management of trigeminal neuralgia [J]. Cleve Clin J Med, 2023, 90(6): 355-362.
[99] Luo YM, Xu JH, Yi ZM, et al. Systematic review and meta-analysis of oxcarbazepine versus carbamazepine for primary trigeminal neuralgia [J]. Chinese Journal of Clinical Pharmacology, 2014(12): 1130-1134.
[100] Razazian N, Baziyar M, Moradian N, et al. Evaluation of the efficacy and safety of pregabalin, venlafaxine, and carbamazepine in patients with painful diabetic peripheral neuropathy: a randomized, double-blind trial [J]. Neurosciences (Riyadh), 2014 19(3): 192-198. PMID: 24983280.
[101] Naderi Y, Rad M, Sadatmoosavi A, et al. Compared to oxcarbazepine and carbamazepine, botulinum toxin type a is a useful therapeutic option for trigeminal neuralgia symptoms: a systematic review [J]. Clin Exp Dent Res, 2024, 10(2): e882.
[102] Li S, Xiang H, Huang YS, et al. Meta-analysis of oxcarbazepine for primary trigeminal neuralgia [J]. Chinese Journal of Gerontology, 2015(24): 7156-7158.
[103] Zhou M, Chen N, He L, et al. Oxcarbazepine for neuropathic pain [J]. Cochrane Database Syst Rev, 2017, 12(12): CD007963.
[104] Waldfogel JM, Nesbit SA, Dy SM, et al. Pharmacotherapy for diabetic peripheral neuropathy pain and quality of life: a systematic review [J]. Neurology, 2017, 88(20): 1958-1967.
[105] Min K, Oh Y, Lee SH, et al. Symptom-based treatment of neuropathic pain in spinal cord-injured patients: a randomized crossover clinical trial [J]. Am J Phys Med Rehabil, 2016, 95(5): 330-338.
[106] Liu X, Wei L, Zeng Q, et al. The treatment of topical drugs for postherpetic neuralgia: a network meta-analysis [J]. Pain Physician, 2020, 23(6): 541-551. PMID: 33185370.
[107] Binder A, Rogers P, Hans G, et al. Impact of topical 5% lidocaine-medicated plasters on sleep and quality of life in patients with postherpetic neuralgia [J]. Pain Manag, 2016, 6(3): 229-239.
[108] Agarwal N, Joshi M. Effectiveness of amitriptyline and lamotrigine in traumatic spinal cord injury-induced neuropathic pain: a randomized longitudinal comparative study [J]. Spinal Cord, 2017, 55(2): 126-130.
[109] Mehta S, Guy S, Lam T, et al. Antidepressants are effective in decreasing neuropathic pain after sci: a meta-analysis [J]. Top Spinal Cord Inj Rehabil, 2015, 21(2): 166-173.
[110] Yasuda H, Hotta N, Kasuga M, et al. Efficacy and safety of 40 mg or 60 mg duloxetine in Japanese adults with diabetic neuropathic pain: results from a randomized, 52-week, open-label study [J]. J Diabetes Investig, 2016, 7(1): 100-108.
[111] Hou S, Huh B, Kim HK, et al. Treatment of chemotherapy induced peripheral neuropathy: systematic review and recommendations [J]. Pain Physician, 2018, 21(6): 571-592. PMID: 30508986.
[112] Wang C, Chen S, Jiang W. Treatment for chemotherapy-induced peripheral neuropathy: a systematic review of randomized control trials [J]. Front Pharmacol, 2022, 13: 1080888.
[113] Wang M, Pei Z, Molassiotis A. Recent advances in managing chemotherapy-induced peripheral neuropathy: a systematic review [J]. Eur J Oncol Nurs, 2022, 58: 102134.
[114] De Liyis BG, Sutedja JC, Tjandra DC, et al. Serotonin norepinephrine reuptake inhibitors in managing neuropathic pain following spinal and non-spinal surgery: a systematic review and meta-analysis of randomized controlled trials [J]. Clin Neurol Neurosurg, 2024, 239: 108223.
[115] Mehta S, McIntyre A, Janzen S, et al. Spinal cord injury rehabilitation evidence team. systematic review of pharmacologic treatments of pain after spinal cord injury: an update [J]. Arch Phys Med Rehabil, 2016, 97(8): 1381-1391, E1.
[116] Mahesh B, Singh VK, Pathak A, et al. Efficacy of duloxetine in patients with central post-stroke pain: a randomized double blind placebo controlled trial [J]. Pain Med, 2023, 24(6): 610-617.
[117] Sommer C, Klose P, Welsch P, et al. Opioids for chronic non-cancer neuropathic pain. An updated systematic review and meta-analysis of efficacy, tolerability and safety in randomized placebo-controlled studies of at least 4 weeks duration [J]. Eur J Pain, 2020, 24(1): 3-18.
[118] Simpson RW, Wlodarczyk JH. Transdermal buprenorphine relieves neuropathic pain: a randomized, double-blind, parallel group, placebo-controlled trial in diabetic peripheral neuropathic pain [J]. Diabetes Care, 2016, 39(9): 1493-1500.
[119] Kim YH, Lee PB, Oh TK. Is magnesium sulfate effective for pain in chronic postherpetic neuralgia patients comparing with ketamine infusion therapy? [J]. J Clin Anesth, 2015, 27(4): 296-300.
[120] Ostrowski H, Roszak J, Komisarek O. Botulinum toxin type A as an alternative way to treat trigeminal neuralgia: a systematic review [J]. Neurol Neurochir Pol, 2019, 53(5): 327-334.
[121] Wei J, Zhu X, Yang G, et al. The efficacy and safety of botulinum toxin type A in treatment of trigeminal neuralgia and peripheral neuropathic pain: a meta-analysis of randomized controlled trials [J]. Brain Behav, 2019, 9(10): E01409.
[122] Rubis A, Juodzbalys G. The use of botulinum toxin A in the management of trigeminal neuralgia: a systematic literature review [J]. J Oral Maxillofac Res, 2020, 11(2): E2.
[123] Hu X, Xia Y, Li J, et al. Efficacy and safety of botulinum toxin type a in the treatment of trigeminal neuralgia: an update on systematic review with meta-analyses [J]. Clin J Pain, 2024, 40(6): 383-392.
[124] Shackleton T, Ram S, Black M, et al. The efficacy of botulinum toxin for the treatment of trigeminal and postherpetic neuralgia: a systematic review with meta-analyses [J]. Oral Surg Oral Med Oral Pathol Oral Radiol, 2016, 122(1): 61-71.
[125] Lin CS, Lin YC, Lao HC, et al. Interventional treatments for postherpetic neuralgia: a systematic review [J]. Pain Physician, 2019, 22(3): 209-228. PMID: 31151330.
[126] Chen L, Zhang Y, Chen Y, et al. Efficacy and safety of botulinum toxin a and pulsed radiofrequency on postherpetic neuralgia: a randomized clinical trial [J]. Contrast Media Mol Imaging, 2022, 2022: 1579937.
[127] Wen B, Wang Y, Zhang C, et al. Efficacy of different interventions for the treatment of postherpetic neuralgia: a Bayesian network meta-analysis [J]. J Int Med Res, 2020, 48(12): 300060520977416.
[128] Li XL, Zeng X, Zeng S, et al. Botulinum toxin A treatment for post-herpetic neuralgia: a systematic review and meta-analysis [J]. Exp Ther Med, 2020, 19(2): 1058-1064.
[129] Wang C, Zhang Q, Wang R, et al. Botulinum toxin type A for diabetic peripheral neuropathy pain: a systematic review and meta-analysis [J]. J Pain Res, 2021, 14: 3855-3863.
[130] Hary V, Schitter S, Martinez V. Efficacy and safety of botulinum A toxin for the treatment of chronic peripheral neuropathic pain: a systematic review of randomized controlled trials and meta-analysis [J]. Eur J Pain, 2022, 26(5): 980-990.
[131] Han ZA, Song DH, Oh HM, et al. Botulinum toxin type A for neuropathic pain in patients with spinal cord injury [J]. Ann Neurol, 2016, 79(4): 569-578.
[132] Mei L, Fengqun M, Zhengyao Z, et al. Efficacy and safety of different drug treatments in patients with spinal-cord injury-related neuropathic pain: a network meta-analysis [J]. Spinal Cord, 2022, 60(11): 943-953.
[133] Yong YL, Tan LT, Ming LC, et al. The Effectiveness and safety of topical capsaicin in postherpetic neuralgia: a systematic review and meta-analysis [J]. Front Pharmacol, 2017, 7: 538.
[134] Derry S, Rice AS, Cole P, et al. Topical capsaicin (high concentration) for chronic neuropathic pain in adults [J]. Cochrane Database Syst Rev, 2017, 1(1): CD007393.
[135] Van Nooten F, Treur M, Pantiri K, et al. Capsaicin 8% patch versus oral neuropathic pain medications for the treatment of painful diabetic peripheral neuropathy: a systematic literature review and network meta-analysis [J]. Clin Ther, 2017, 39(4): 787-803, E18.
[136] Goodwin B, Chiplunkar M, Salerno R, et al. Topical capsaicin for the management of painful diabetic neuropathy: a narrative systematic review [J]. Pain Manag, 2023, 13(5): 309-316.
[137] Simpson DM, Robinson-Papp J, Van J, et al. Capsaicin 8% patch in painful diabetic peripheral neuropathy: a randomized, double-blind, placebo-controlled study [J]. J Pain, 2017, 18(1): 42-53.
[138] Cabezón-Gutiérrez L, Custodio-Cabello S, Palka-Kotlowska M, et al. High-dose 8% capsaicin patch in treatment of chemotherapy-induced peripheral neuropathy. a systematic review [J]. J Pain Symptom Manage, 2020, 60(5): 1047-1054, E1.
[139] Olusanya A, Yearsley A, Brown N, et al. Capsaicin 8% patch for spinal cord injury focal neuropathic pain, a randomized controlled trial [J]. Pain Med, 2023, 24(1): 71-78.
[140] Liu PH, Yuan D, Zhao YQ, et al. Systematic review of lamotrigine monotherapy for trigeminal neuralgia [J]. Chinese Remedies & Clinics, 2016, 16(4): 482-485.
[141] Liampas A, Velidakis N, Georgiou T, et al. Prevalence and management challenges in central post-stroke neuropathic pain: a systematic review and meta-analysis [J]. Adv Ther, 2020, 37(7): 3278-3291.
[142] Peng HR, Li S, Huang YS, et al. Systematic review of vaccinia virus-inflamed rabbit skin extract for postherpetic neuralgia [J]. Modern Preventive Medicine, 2015, 42(11): 2110-2112. (In Chinese)
[143] Zhang Y, Hu ZS, Tu Y, et al. Meta-analysis of vaccinia vaccination inflammation skin extract for diabetic peripheral neuropathy [J]. Anhui Medical and Pharmaceutical Journal, 2022, 26(1): 1-5.
[144] Finnerup NB, Attal N, Haroutounian S, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis [J]. Lancet Neurol, 2015, 14(2): 162-173.
[145] Moisset X, Bouhassira D, Avez Couturier J, et al. Pharmacological and non-pharmacological treatments for neuropathic pain: Systematic review and French recommendations [J]. Rev Neurol (Paris), 2020, 176(5): 325-352.
[146] Bouchenaki H, Bégou M, Magy L, et al. Pharmacological management of neuropathic pain [J]. Therapie, 2019, 74(6): 633-643.
[147] Mu A, Weinberg E, Moulin DE, et al. Pharmacologic management of chronic neuropathic pain: review of the Canadian pain society consensus statement [J]. Can Fam Physician, 2017, 63(11): 844-852. PMID: 29138154.
[148] Neuropathic Pain Expert Group of National Pain Management Quality Control Center. Chinese guidelines for assessment and management of neuropathic pain (2024 edition) [J]. Chinese Journal of Pain Medicine, 2024, 30(1): 5-14.
[149] Gilron I, Tu D, Holden RR, et al. Combination of morphine with nortriptyline for neuropathic pain [J]. Pain, 2015, 156(8): 1440-1448.
[150] Pain and Sensory Disorders Committee of Chinese Neurologist Association. Expert consensus on diagnosis and treatment of diabetic peripheral neuropathic pain [J]. Clinical Education of General Practice, 2019, 17(2): 100-103, 107.
[151] Oliveira RAA, Baptista AF, Sá KN, et al. Clinicians participants of the panel of experts recommended by the Brazilian Academy of Neurology; Andrade DC. Pharmacological treatment of central neuropathic pain: consensus of the Brazilian Academy of Neurology [J]. Arq Neuropsiquiatr, 2020, 78(11): 741-752.
[152] Jiang X, Yan W, Wan R, et al. Effects of repetitive transcranial magnetic stimulation on neuropathic pain: a systematic review and meta-analysis [J]. Neurosci Biobehav Rev, 2022, 132: 130-141.
[153] Gao C, Zhu Q, Gao Z, et al. Can noninvasive brain stimulation improve pain and depressive symptoms in patients with neuropathic pain? a systematic review and meta-analysis [J]. J Pain Symptom Manage, 2022, 64(4): E203-E215.
[154] Zhang KL, Yuan H, Wu FF, et al. Analgesic effect of noninvasive brain stimulation for neuropathic pain patients: a systematic review [J]. Pain Ther, 2021, 10(1): 315-332.
[155] André-Obadia N, Hodaj H, Hodaj E, et al. Better fields or currents? A head-to-head comparison of transcranial magnetic (rtms) versus direct current stimulation (tdcs) for neuropathic pain [J]. Neurotherapeutics, 2023, 20(1): 207-219.
[156] David MCMM, Moraes AA, Costa MLD, et al. Transcranial direct current stimulation in the modulation of neuropathic pain: a systematic review [J]. Neurol Res, 2018, 40(7): 555-563.
[157] Zhang YH, Hu HY, Xiong YC, et al. Exercise for neuropathic pain: a systematic review and expert consensus [J]. Front Med (Lausanne), 2021, 8: 756940.
[158] Holmes A, Chang YP. Non-pharmacological management of neuropathic pain in older adults: a systematic review [J]. Pain Med, 2024, 25(1): 47-56.
[159] Ibarra AMC, Biasotto-Gonzalez DA, Kohatsu EYI, et al. Photobiomodulation on trigeminal neuralgia: systematic review [J]. Lasers Med Sci, 2021, 36(4): 715-722.
[160] Korada HY, Arora E, Maiya GA, et al. Effectiveness of photobiomodulation therapy on neuropathic pain, nerve conduction and plantar pressure distribution in diabetic peripheral neuropathy: a systematic review [J]. Curr Diabetes Rev, 2023, 19(9): E290422204244.
[161] Sæbø H, Naterstad IF, Joensen J, et al. Pain and disability of conservatively treated distal radius fracture: a triple-blinded randomized placebo-controlled trial of photobiomodulation therapy [J]. Photobiomodul Photomed Laser Surg, 2022, 40(1): 33-41.
[162] Haghighat S, Rezazadeh F, Sedarat H, et al. Efficacy of laser therapy in trigeminal neuralgia: a systematic review [J]. J Dent (Shiraz), 2024, 25(1): 17-25.
[163] Al-Azab IM, Abo Elyazed TI, El Gendy AM, et al. Effect of electromagnetic therapy versus low-level laser therapy on diabetic patients with trigeminal neuralgia: a randomized control trial [J]. Eur J Phys Rehabil Med, 2023, 59(2): 183-191.
[164] De Pedro M, López-Pintor RM, de la Hoz-Aizpurua JL, et al. Efficacy of low-level laser therapy for the therapeutic management of neuropathic orofacial pain: a systematic review [J]. J Oral Facial Pain Headache, 2020, 34(1): 13-30.
[165] MA, Ummer VS, Maiya AG, et al. Low level laser therapy for the patients with painful diabetic peripheral neuropathy- a systematic review [J]. Diabetes Metab Syndr, 2019, 13(4): 2667-2670.
[166] Ahmed I, Bandpei MAM, Gilani SA, et al. Effectiveness of low-level laser therapy in patients with discogenic lumbar radiculopathy: a double-blind randomized controlled trial [J]. J Healthc Eng, 2022, 2022: 6437523.
[167] Motwani M, Fadnavis A, Dhole A. Efficacy of transcutaneous electrical nerve stimulation (TENS) in the management of trigeminal neuralgia: a systematic review and meta-analysis [J]. J Clin Exp Dent, 2023, 15(6): E505-E510.
[168] Zheng Y, Liu CW, Hui Chan DX, et al. Neurostimulation for chronic pain: a systematic review of high-quality randomized controlled trials with long-term follow-up [J]. Neuromodulation, 2023, 26(7): 1276-1294.
[169] Ing MR, Hellreich PD, Johnson DW, et al. Transcutaneous electrical nerve stimulation for chronic post-herpetic neuralgia [J]. Int J Dermatol, 2015, 54(4): 476-480.
[170] Eid MM, Hamed NS, Abdelbasset WK, et al. A comparative study between transcutaneous electrical nerve stimulation and pulsed electromagnetic field therapy in the management of post-herpetic neuralgia of the sciatic nerve [J]. Medicine (Baltimore), 2022, 101(44): E31433.
[171] Upton GA, Tinley P, Al-Aubaidy H, et al. The influence of transcutaneous electrical nerve stimulation parameters on the level of pain perceived by participants with painful diabetic neuropathy: a crossover study [J]. Diabetes Metab Syndr, 2017, 11(2): 113 118.
[172] Bernetti A, Agostini F, de Sire A, et al. Neuropathic pain and rehabilitation: a systematic review of international guidelines [J]. Diagnostics (Basel), 2021, 11(1): 74.
[173] Gewandter JS, Culakova E, Davis JN, et al. Wireless transcutaneous electrical nerve stimulation (tens) for chronic chemotherapy-induced peripheral neuropathy (cipn): a proof-of-concept randomized clinical trial [J]. J Pain, 2024, 25(5): 104431.
[174] Klafke N, Bossert J, Kröger B, et al. Prevention and treatment of chemotherapy-induced peripheral neuropathy (cipn) with non pharmacological interventions: clinical recommend-ations from a systematic scoping review and an expert consensus process [J]. Med Sci (Basel), 2023, 11(1): 15.
[175] Tilak M, Isaac SA, Fletcher J, et al. Mirror therapy and transcutaneous electrical nerve stimulation for management of phantom limb pain in amputees- a single blinded randomized controlled trial [J]. Physiother Res Int, 2016, 21(2): 109-115.
[176] Luyao H, Xiaoxiao Y, Tianxiao F, et al. Management of cervical spondylotic radiculopathy: a systematic review [J]. Global Spine J, 2022, 12(8): 1912-1924.
[177] Yang Y, Tang Y, Qin H, et al. Efficacy of transcutaneous electrical nerve stimulation in people with pain after spinal cord injury: a meta-analysis [J]. Spinal Cord, 2022, 60(5): 375-381.
[178] Karri J, Marathe A, Smith TJ, et al. The use of scrambler therapy in treating chronic pain syndromes: a systematic review [J]. Neuromodulation, 2023, 26(8): 1499-1509.
[179] Pachman DR, Weisbrod BL, Seisler DK, et al. Pilot evaluation of scrambler therapy for the treatment of chemotherapy-induced peripheral neuropathy [J]. Support Care Cancer, 2015, 23(4): 943-951.
[180] Childs DS, Le-Rademacher JG, McMurray R, et al. Randomized trial of scrambler therapy for chemotherapy-induced peripheral neuropathy: crossover analysis [J]. J Pain Symptom Manage, 2021, 61(6): 1247-1253.
[181] Chen L, Qing A, Zhu T, et al. Effect and safety of extracorporeal shockwave therapy for postherpetic neuralgia: a randomized single blind clinical study [J]. Front Neurol, 2022, 13: 948024.
[182] Lee SH, Ryu KH, Kim PO, et al. Efficacy of extracorporeal shockwave therapy in the treatment of postherpetic neuralgia: a pilot study [J]. Medicine (Baltimore), 2020, 99(12): e19516.
[183] Wu HY, Wu JH, Wen GW. Meta-analysis of the clinical efficacy of extracorporeal shock wave therapy for cervical spondylotic radiculopathy [J]. Clinical Journal of Chinese Medicine, 2023, 15(11): 132-137.
[184] Säisänen L, Huttunen J, Hyppönen J, et al. Efficacy and tolerability in patients with chronic facial pain of two consecutive treatment periods of rTMS applied over the facial motor cortex, using protocols differing in stimulation frequency, duration, and train pattern [J]. Neurophysiol Clin, 2022, 52(2): 95-108.
[185] Ma SM, Ni JX, Li XY, et al. High-frequency repetitive transcranial magnetic stimulation reduces pain in postherpetic neuralgia [J]. Pain Med, 2015, 16(11): 2162-2170.
[186] Dai Q, Xu A, Wang K, et al. The efficacy of repetitive transcranial magnetic stimulation in postherpetic neuralgia: a meta-analysis of randomized controlled trials [J]. Front Neurol, 2024, 15: 1365445.
[187] Liampas A, Rekatsina M, Vadalouca A, et al. Non pharmacological management of painful peripheral neuropathies: a systematic review [J]. Adv Ther, 2020, 37(10): 4096-4106.
[188] Yang S, Kwak SG, Choi GS, et al. Short-term effect of repetitive transcranial magnetic stimulation on diabetic peripheral neuropathic pain [J]. Pain Physician, 2022, 25(2): E203-E209. PMID: 35322973.
[189] Pei Q, Wu B, Tang Y, et al. Repetitive transcranial magnetic stimulation at different frequencies for postherpetic neuralgia: a double-blind, sham-controlled, randomized trial [J]. Pain Physician, 2019, 22(4): E303-E313. PMID: 31337172.
[190] Zeng H, Pacheco-Barrios K, Cao Y, et al. Non-invasive neuromodulation effects on painful diabetic peripheral neuropathy: a systematic review and meta-analysis [J]. Sci Rep, 2020, 10(1): 19184.
[191] Bonifácio de Assis ED, Martins WKN, et al. Effects of rTMS and tDCS on neuropathic pain after brachial plexus injury: a randomized placebo-controlled pilot study [J]. Sci Rep, 2022, 12(1): 1440.
[192] Garcia-Pallero MÁ, Cardona D, Rueda-Ruzafa L, et al. Central nervous system stimulation therapies in phantom limb pain: a systematic review of clinical trials [J]. Neural Regen Res, 2022, 17(1): 59-64.
[193] Aamir A, Girach A, Sarrigiannis PG, et al. Repetitive magnetic stimulation for the management of peripheral neuropathic pain: a systematic review [J]. Adv Ther, 2020, 37(3): 998-1012.
[194] Attal N, Ayache SS, Ciampi De Andrade D, et al. Repetitive transcranial magnetic stimulation and transcranial direct-current stimulation in neuropathic pain due to radiculopathy: a randomized sham-controlled comparative study [J]. Pain, 2016, 157(6): 1224-1231.
[195] Saleh C, Ilia TS, Jaszczuk P, et al. Is transcranial magnetic stimulation as treatment for neuropathic pain in patients with spinal cord injury efficient? a systematic review [J]. Neurol Sci, 2022, 43(5): 3007-3018.
[196] Gao F, Chu H, Li J, et al. Repetitive transcranial magnetic stimulation for pain after spinal cord injury: a systematic review and meta-analysis [J]. J Neurosurg Sci, 2017, 61(5): 514-522.
[197] Li L, Huang H, Yu Y, et al. Non-invasive brain stimulation for neuropathic pain after spinal cord injury: a systematic review and network meta-analysis [J]. Front Neurosci, 2022, 15: 800560.
[198] Zhang RG, Wang FY, Zhang JQ, et al. Systematic review of repetitive transcranial magnetic stimulation for improving neuropathic pain in patients with spinal cord injury [J]. Chinese Journal of Physical Medicine and Rehabilitation, 2021, 43(7): 645-649.
[199] Wen BB, Dai X, Wang XX, et al. Systematic review and meta-analysis of repetitive transcranial magnetic stimulation for neuropathic pain after spinal cord injury [J]. The Journal of Cervicodynia and Lumbodynia, 2024, 45(1): 124-128.
[200] Gurdiel-Álvarez F, Navarro-López V, Varela-Rodríguez S, et al. Transcranial magnetic stimulation therapy for central post-stroke pain: systematic review and meta-analysis [J]. Front Neurosci, 2024, 18: 1345128.
[201] Mayor RS, Ferreira NR, Lanzaro C, et al. Noninvasive transcranial brain stimulation in central post-stroke pain: a systematic review [J]. Scand J Pain, 2024, 24(1).
[202] Lizi H, Jiaojiao K, Dan W, et al. Non-invasive brain stimulation improves pain in patients with central post-stroke pain: a systematic review and meta-analysis [J]. Top Stroke Rehabil, 2024, 3: 1-16.
[203] Chen CC, Chuang YF, Huang AC, et al. The antalgic effects of non-invasive physical modalities on central post-stroke pain: a systematic review [J]. J Phys Ther Sci, 2016, 28(4): 1368-1373.
[204] Liu Y, Miao R, Zou H, et al. Repetitive transcranial magnetic stimulation in central post-stroke pain: a meta-analysis and systematic review of randomized controlled trials [J]. Front Neurosci, 2024, 18: 1367649.
[205] Alipour A, Mohammadi R. Evaluation of the separate and combined effects of anodal tDCS over the M1 and F3 regions on pain relief in patients with type-2 diabetes suffering from neuropathic pain [J]. Neurosci Lett, 2024, 818: 137554.
[206] Mehta S, McIntyre A, Guy S, et al. Effectiveness of transcranial direct current stimulation for the management of neuropathic pain after spinal cord injury: a meta-analysis [J]. Spinal Cord, 2015, 53(11): 780-785.
[207] Yeh NC, Yang YR, Huang SF, et al. Effects of transcranial direct current stimulation followed by exercise on neuropathic pain in chronic spinal cord injury: a double-blinded randomized controlled pilot trial [J]. Spinal Cord, 2021, 59(6): 684-692.
[208] Li R, Liu Y, Xue R, et al. Effectiveness of nonpharmacologic interventions for chemotherapy-induced peripheral neuropathy in patients with breast cancer: a systematic review and network meta-analysis [J]. Cancer Nurs, 2023.
[209] Elshinnawy AM, Eraky ZS, Abdelaziz SS, et al. Effect of cold application versus transcutaneous nerve stimulation on chemotherapy induced diabetic peripheral neuropathy post mastectomy [J]. Physiother Res Int, 2024, 29(1): E2051.
[210] Robinson CC, Barreto RPG, Plentz RDM. Effects of whole body vibration in individuals with diabetic peripheral neuropathy: a systematic review [J]. J Musculoskelet Neuronal Interact, 2018, 18(3): 382-388.
[211] Kessler NJ, Lockard MM, Fischer J. Whole body vibration improves symptoms of diabetic peripheral neuropathy [J]. J Bodyw Mov Ther, 2020, 24(2): 1-3.
[212] Wong ML, Widerstrom-Noga E, Field-Fote EC. Effects of whole-body vibration on neuropathic pain and the relationship between pain and spasticity in persons with spinal cord injury [J]. Spinal Cord, 2022, 60(11): 963-970.
[213] Knoerl R, Giobbie-Hurder A, Berfield J, et al. Yoga for chronic chemotherapy-induced peripheral neuropathy pain: a pilot, randomized controlled trial [J]. J Cancer Surviv, 2022, 16(4): 882-891.
[214] Yildirim P, Gultekin A. The effect of a stretch and strength-based yoga exercise program on patients with neuropathic pain due to lumbar disc herniation [J]. Spine (Phila Pa 1976), 2022, 47(10): 711-719.
[215] Bakr SM, Knight JA, Shlobin NA, et al. Spinal cord stimulation for treatment of chronic neuropathic pain in adolescent patients: a single-institution series, systematic review, and individual participant data meta-analysis [J]. Neurosurg Focus, 2022, 53(4): E13.
[216] Duarte RV, Nevitt S, McNicol E, et al. Systematic review and meta-analysis of placebo/sham controlled randomised trials of spinal cord stimulation for neuropathic pain [J]. Pain, 2020, 161(1): 24-35.
[217] Wang D, Lu Y, Han Y, et al. The influence of etiology and stimulation target on the outcome of deep brain stimulation for chronic neuropathic pain: a systematic review and meta-analysis [J]. Neuromodulation, 2024, 27(1): 83-94.
[218] Yang R, Xiong B, Wang M, et al. Gamma knife surgery and deep brain stimulation of the centromedian nucleus for chronic pain: a systematic review [J]. Asian J Surg, 2023, 46(9): 3437-3446.
[219] Aggarwal A, Suresh V, Gupta B, et al. Post-herpetic neuralgia: a systematic review of current interventional pain management strategies [J]. J Cutan Aesthet Surg, 2020, 13(4): 265-274.
[220] Wang C, Yuan F, Cai L, et al. Ultrasound-guided stellate ganglion block combined with extracorporeal shock wave therapy on postherpetic neuralgia [J]. J Healthc Eng, 2022, 2022: 9808994.
[221] Guo Y, Ou CH. Systematic review of early regional nerve block for prevention and treatment of herpes zoster neuralgia [J]. Medical Journal of National Defending Forces in Southwest China, 2019, 29(10): 1021-1024.
[222] Mu Q, Chai JL, Lyu HL, et al. Meta-analysis of clinical efficacy and safety of stellate ganglion block for postherpetic neuralgia [J]. China Medical Herald, 2022, 19(19): 103-106. (In Chinese).
[223] Pain Medicine Branch of Chinese Medical Association. Expert consensus on the treatment of degenerative spinal radicular pain [J]. Chinese Medical Journal, 2019, 99(15): 1133-1137.
[224] Chen J, Feng X, Zeng C, et al. Meta-analysis of ultrasound-guided cervical nerve root block for cervical spondylotic radiculopathy [J]. Chinese Journal of Painology, 2022, 18(2): 265-272.
[225] Wu Z, Zhao Y, Liu J, et al. Comparison of the safety and efficacy of radiofrequency thermocoagulation with percutaneous balloon compression for treating trigeminal neuralgia: a systematic review and meta-analysis [J]. Front Neurol, 2023, 14: 1178335.
[226] Wu J, Xiao Y, Chen B, et al. Efficacy and safety of microvascular decompression versus percutaneous balloon compression in the treatment of trigeminal neuralgia: a systematic review and meta-analysis [J]. Ann Palliat Med, 2022, 11(4): 1391-1400.
[227] Zhou Y, Dou NN, Liu XL, et al. A comparative review of the outcome following mvd and pbc in patients with trigeminal neuralgia [J]. J Neurol Surg A Cent Eur Neurosurg, 2023, 84(5): 470-476.
[228] Fan X, Fu Z, Ma K, et al. Chinese expert consensus on minimally invasive interventional treatment of trigeminal neuralgia [J]. Front Mol Neurosci, 2022, 15: 953765.
[229] Wang KX, He ZX, Yang XC, et al. Comparison of efficacy and safety between radiofrequency thermocoagulation and balloon compression for trigeminal neuralgia: A meta-analysis [J]. Chinese Journal of Painology, 2023, 19(6): 941-950.
[230] Wu XK, Lian YJ, Chen Y, et al. Meta-analysis comparing the efficacy and safety of percutaneous radiofrequency thermocoagulation versus balloon compression for primary trigeminal neuralgia [J]. Chinese Journal of Painology, 2023, 19(1): 36-43.
[231] Ren YE, Liu XH, Cheng ZX, et al. Chinese expert consensus on percutaneous balloon compression for trigeminal neuralgia (2022 edition) [J]. Chinese Journal of Painology, 2022, 18(4): 437 448.
[232] Functional Neurosurgery Expert Committee of Chinese Medical Doctor Association, Functional Neurosurgery Expert Committee of World Chinese Neurosurgical Association, Neurosurgery Professional Committee of Chinese Research Hospital Association, et al. Chinese expert consensus on percutaneous balloon compression for trigeminal neuralgia [J]. Chinese Journal of Brain Diseases and Rehabilitation (Electronic Edition), 2022, 12(5): 260-268.
[233] Orhurhu V, Khan F, Quispe RC, et al. Use of radiofrequency ablation for the management of facial pain: a systematic review [J]. Pain Physician, 2020, 23(6): E559-E580. PMID: 33185371.
[234] Garcia-Isidoro S, Castellanos-Sanchez VO, Iglesias-Lopez E, et al. Invasive and non-invasive electrical neuromodulation in trigeminal nerve neuralgia: a systematic review and meta-analysis [J]. Curr Neuropharmacol, 2021, 19(3): 320-333.
[235] Abbas DN, Reyad RM. Thermal versus super voltage pulsed radiofrequency of stellate ganglion in post-mastectomy neuropathic pain syndrome: a prospective randomized trial [J]. Pain Physician, 2018, 21(4): 351-362. PMID: 30045592.
[236] Gupta H, Vance C, Bansal V, et al. A narrative review of pulsed radiofrequency for the treatment of carpal tunnel syndrome [J]. Pain Pract, 2024, 24(2): 374-382.
[237] Wang C, Dou Z, Yan M, et al. Efficacy and safety of pulsed radiofrequency in herpes zoster related trigeminal neuralgia: a systematic review and meta-analysis [J]. J Pain Res, 2023, 16: 341-355.
[238] Qiu ZQ, Zhong XK, Yang QM, et al. Network meta-analysis of the efficacy and safety of different treatment regimens for drug-resistant trigeminal neuralgia [J]. Journal of Clinical Neurology, 2023, 36(2): 90-98.
[239] Shi Y, Wu W. Treatment of neuropathic pain using pulsed radiofrequency: a meta-analysis [J]. Pain Physician, 2016, 19(7): 429-444. PMID: 27676660.
[240] Makharita MY, El Bendary HM, et al. Ultrasound-guided pulsed radiofrequency in the management of thoracic postherpetic neuralgia: a randomized, double-blinded, controlled trial [J]. Clin J Pain, 2018, 34(11): 1017-1024.
[241] Wu CY, Lin HC, Chen SF, et al. Efficacy of pulsed radiofrequency in herpetic neuralgia: a meta-analysis of randomized controlled trials [J]. Clin J Pain, 2020, 36(11): 887-895.
[242] Tian YF. Systematic review of the clinical efficacy of pulsed radiofrequency therapy for postherpetic neuralgia [J]. Smart Healthcare, 2023, 9(16): 105-108.
[243] Hetta DF, Mohamed SAB, Mohamed KH, et al. Pulsed radiofrequency on thoracic dorsal root ganglion versus thoracic paravertebral nerve for chronic postmastectomy pain, a randomized trial: 6-month results [J]. Pain Physician, 2020, 23(1): 23-35. PMID: 32013276.
[244] Marliana A, Setyopranoto I, Setyaningsih I, et al. The effect of pulsed radiofrequency on radicular pain in lumbal herniated nucleus pulposus: a systematic review and meta-analysis [J]. Anesth Pain Med, 2021, 11(2): E111420.
[245] Kwak SG, Lee DG, Chang MC. Effectiveness of pulsed radiofrequency treatment on cervical radicular pain: a meta-analysis [J]. Medicine (Baltimore), 2018, 97(31): E11761.
[246] Park S, Park JH, Jang JN, et al. Pulsed radiofrequency of lumbar dorsal root ganglion for lumbar radicular pain: a systematic review and meta-analysis [J]. Pain Pract, 2024, 24(5): 772-785.
[247] Erken B, Edipoglu IS. Efficacy of high-voltage pulsed radiofrequency of the dorsal root ganglion for treatment of chronic lumbosacral radicular pain: a randomized clinical trial [J]. Neuromodulation, 2024, 27(1): 135-140.
[248] Wu H, Zhou J, Chen J, et al. Therapeutic efficacy and safety of radiofrequency ablation for the treatment of trigeminal neuralgia: a systematic review and meta-analysis [J]. J Pain Res, 2019, 12: 423-441.
[249] Zhang X, Peng L, Liu D. Radiofrequency therapies for trigeminal neuralgia: a systematic review and updated meta-analysis [J]. Pain Physician, 2022, 25(9): E1327-E1337. PMID: 36608005.
[250] Abdel-Rahman KA, Elawamy AM, Mostafa MF, et al. Combined pulsed and thermal radiofrequency versus thermal radiofrequency alone in the treatment of recurrent trigeminal neuralgia after microvascular decompression: a double blinded comparative study [J]. Eur J Pain, 2020, 24(2): 338-345.
[251] Li X, Chen P, He J, et al. Comparison of the efficacy and safety of temporary spinal cord stimulation versus pulsed radiofrequency for postherpetic neuralgia: a prospective randomized controlled trial [J]. Pain Res Manag, 2022, 2022: 3880424.
[252] Xue S, Yang WJ, Cao ZX, et al. Comparing the efficacy and safety of short-term spinal cord stimulation and pulsed radiofrequency for zoster-related pain: a systematic review and meta-analysis [J]. Medicine (Baltimore), 2022, 101(11): e29073.
[253] Sheng L, Liu Z, Zhou W, et al. Short-term spinal cord stimulation or pulsed radiofrequency for elderly patients with postherpetic neuralgia: a prospective randomized controlled trial [J]. Neural Plast, 2022, 2022: 7055697.
[254] Pain Physician Branch of Chinese Medical Doctor Association, Neuromodulation Committee of Chinese Medical Doctor Association. Chinese expert consensus on percutaneous short-term nerve electrical stimulation for herpes zoster neuralgia [J]. Chinese Journal of Pain Medicine, 2021, 27(11): 801-805.
[255] Raghu ALB, Parker T, Aziz TZ, et al. Invasive electrical neuromodulation for the treatment of painful diabetic neuropathy: systematic review and meta-analysis [J]. Neuromodulation, 2021, 24(1): 13-21.
[256] Henson JV, Varhabhatla NC, Bebic Z, et al. Spinal cord stimulation for painful diabetic peripheral neuropathy: a systematic review [J]. Pain Ther, 2021, 10(2): 895-908.
[257] D'Souza RS, Langford B, Dombovy-Johnson M, et al. Neuromodulation interventions for the treatment of painful diabetic neuropathy: a systematic review [J]. Curr Pain Headache Rep, 2022, 26(5): 365-377.
[258] Hoelzer BC, Edgar D, Lu SP, et al. Indirect comparison of 10 khz spinal cord stimulation (scs) versus traditional low-frequency scs for the treatment of painful diabetic neuropathy: a systematic review of randomized controlled trials [J]. Biomedicines, 2022, 10(10): 2630.
[259] Duarte RV, Nevitt S, Maden M, et al. Spinal cord stimulation for the management of painful diabetic neuropathy: a systematic review and meta-analysis of individual patient and aggregate data [J]. Pain, 2021, 162(11): 2635-2643.
[260] Xu L, Sun Z, Casserly E, et al. Advances in interventional therapies for painful diabetic neuropathy: a systematic review [J]. Anesth Analg, 2022, 134(6): 1215-1228.
[261] D'Souza RS, Her YF, Jin MY, et al. Neuromodulation therapy for chemotherapy-induced peripheral neuropathy: a systematic review [J]. Biomedicines, 2022, 10(8): 1909.
[262] Fatima K, Javed SO, Saleem A, et al. Long-term efficacy of spinal cord stimulation for chronic primary neuropathic pain in the contemporary era: a systematic review and meta-analysis [J]. J Neurosurg Sci, 2024, 68(1): 128-139.
[263] Giammalva GR, Paolini F, Bonosi L, et al. Spinal cord stimulation meets them all: an effective treatment for different pain conditions. our experience and literature review [J]. Acta Neurochir Suppl, 2023, 135: 179-195.
[264] Grider JS, Manchikanti L, Carayannopoulos A, et al. Effectiveness of spinal cord stimulation in chronic spinal pain: a systematic review [J]. Pain Physician, 2016, 19(1): E33-E54. PMID: 26752493.
[265] Kapural L, Peterson E, Provenzano DA, et al. Clinical evidence for spinal cord stimulation for failed back surgery syndrome (fbss): systematic review [J]. Spine (Phila Pa 1976), 2017, 42(Suppl 14): S61-S66.
[266] Dombovy-Johnson ML, Hunt CL, Morrow MM, et al. Current evidence lacking to guide clinical practice for spinal cord stimulation in the treatment of neuropathic pain in spinal cord injury: a review of the literature and a proposal for future study [J]. Pain Pract, 2020, 20(3): 325-335.
[267] Galafassi GZ, Simm Pires de Aguiar PH, Simm RF, et al. Neuromodulation for medically refractory neuropathic pain: spinal cord stimulation, deep brain stimulation, motor cortex stimulation, and posterior insula stimulation [J]. World Neurosurg, 2021, 146: 246-260.
[268] Akyuz G, Kuru P. Systematic review of central post stroke pain: what is happening in the central nervous system? [J]. Am J Phys Med Rehabil, 2016, 95(8): 618-627.
[269] Mo JJ, Hu WH, Zhang C, et al. Motor cortex stimulation: a systematic literature-based analysis of effectiveness and case series experience [J]. BMC Neurol, 2019, 19(1): 48.
[270] Gunduz ME, Pacheco-Barrios K, Bonin Pinto C, et al. Effects of combined and alone transcranial motor cortex stimulation and mirror therapy in phantom limb pain: a randomized factorial trial [J]. Neurorehabil Neural Repair, 2021, 35(8): 704-716.
[271] Xu XM, Luo H, Rong BB, et al. Nonpharmacological therapies for central poststroke pain: a systematic review [J]. Medicine (Baltimore), 2020, 99(42): E22611.
[272] Ni Y, Yang L, Han R, et al. Implantable peripheral nerve stimulation for trigeminal neuropathic pain: a systematic review and meta-analysis [J]. Neuromodulation, 2021, 24(6): 983-991.
[273] Sarica C, Iorio-Morin C, Aguirre-Padilla DH, et al. Clinical outcomes and complications of peripheral nerve field stimulation in the management of refractory trigeminal pain: a systematic review and meta-analysis [J]. J Neurosurg, 2022, 137(5): 1387-1395.
[274] Char S, Jin MY, Francio VT, et al. Implantable peripheral nerve stimulation for peripheral neuropathic pain: a systematic review of prospective studies [J]. Biomedicines, 2022, 10(10): 2606.
[275] Gilmore C, Ilfeld B, Rosenow J, et al. Percutaneous peripheral nerve stimulation for the treatment of chronic neuropathic postamputation pain: a multicenter, randomized, placebo controlled trial [J]. Reg Anesth Pain Med, 2019, 44(6): 637-645.
[276] Xu J, Sun Z, Wu J, et al. Peripheral nerve stimulation in pain management: a systematic review [J]. Pain Physician, 2021, 24(2): E131-E152. PMID: 33740342.
[277] Smith BJ, Twohey EE, Dean KP, et al. Peripheral nerve stimulation for the treatment of postamputation pain: a systematic review [J]. Am J Phys Med Rehabil, 2023, 102(9): 846-854.
[278] Piedade GS, Gillner S, McPhillips PS, et al. Frequency dependency of therapeutic efficacy in dorsal root ganglion stimulation for neuropathic pain [J]. Acta Neurochir (Wien), 2022, 164(4): 1193-1199.
[279] Kumru H, Benito-Penalva J, Kofler M, et al. Analgesic effect of intrathecal baclofen bolus on neuropathic pain in spinal cord injury patients [J]. Brain Res Bull, 2018, 140: 205-211.
[280] Holste K, Chan AY, Rolston JD, et al. Pain outcomes following microvascular decompression for drug-resistant trigeminal neuralgia: a systematic review and meta-analysis [J]. Neurosurgery, 2020, 86(2): 182-190.
[281] Di Carlo DT, Benedetto N, Marani W, et al. Microvascular decompression for trigeminal neuralgia due to vertebrobasilar artery compression: a systematic review and meta-analysis [J]. Neurosurg Rev, 2022, 45(1): 285-294.
[282] Jiao L, Ye H, Lv J, et al. A systematic review of repeat microvascular decompression for recurrent or persistent trigeminal neuralgia [J]. World Neurosurg, 2022, 158: 226-233.
[283] Sun J, Wang M, Zhang L, et al. A meta-analysis of the effectiveness and safety of microvascular decompression in elderly patients with trigeminal neuralgia [J]. J Clin Neurosci, 2022, 99: 22-34.
[284] Di Carlo DT, Benedetto N, Perrini P. Clinical outcome after microvascular decompression for trigeminal neuralgia: a systematic review and meta-analysis [J]. Neurosurg Rev, 2022, 46(1): 8.
[285] Chen L, Shang Y, Zhang Y, et al. Endoscopic microvascular decompression versus microscopic microvascular decompression for trigeminal neuralgia: a systematic review and meta-analysis [J]. J Clin Neurosci, 2023, 117: 73-78.
[286] Tuleasca C, Régis J, Sahgal A, et al. Stereotactic radiosurgery for trigeminal neuralgia: a systematic review [J]. J Neurosurg, 2019, 130(3): 733-757.
[287] De La Peña NM, Singh R, Anderson ML, et al. High-dose frameless stereotactic radiosurgery for trigeminal neuralgia: a single-institution experience and systematic review [J]. World Neurosurg, 2022, 167: E432-E443.
[288] Spina A, Nocera G, Boari N, et al. Efficacy of gamma knife radiosurgery in the management of multiple sclerosis-related trigeminal neuralgia: a systematic review and meta-analysis [J]. Neurosurg Rev, 2021, 44(6): 3069-3077.
[289] Radioneurosurgery Committee of World Chinese Neurosurgical Society. Chinese expert consensus on gamma knife radiosurgery for trigeminal neuralgia (2020 edition) [J]. Chinese Journal of Neurosurgery, 2020, 36(10): 984-989.
[290] Li ZH, Zhang J, Chen YH, et al. Meta-analysis of the efficacy and safety of microvascular decompression for glossopharyngeal neuralgia [J]. Chinese Journal of Neurosurgery, 2019, 35(2): 197-203.
[291] Siempis T, Rehder R, Voulgaris S, et al. Stereotactic radiosurgery for idiopathic glossopharyngeal neuralgia: a systematic review [J]. World Neurosurg X, 2024, 22: 100325.
[292] Berckemeyer MA, Suarez-Meade P, Carcelen MFV, et al. Current advances in the surgical treatment of glossopharyngeal neuralgia [J]. Neurosurg Rev, 2023, 46(1): 47.
[293] Lu VM, Goyal A, Graffeo CS, et al. Glossopharyngeal neuralgia treatment outcomes after nerve section, microvascular decompression, or stereotactic radiosurgery: a systematic review and meta-analysis [J]. World Neurosurg, 2018, 120: 572-582, E7.
[294] Kaye J, Daggubati LC, Zeller S, et al. Repeat gamma knife radiosurgery for recurrent glossopharyngeal neuralgia: a systematic review and our initial experience [J]. Stereotact Funct Neurosurg, 2020, 98(5): 324-330.
[295] Best TJ, Best CA, Best AA, et al. Surgical peripheral nerve decompression for the treatment of painful diabetic neuropathy of the foot- A level 1 pragmatic randomized controlled trial [J]. Diabetes Res Clin Pract, 2019, 147: 149-156.
[296] Fadel ZT, Imran WM, Azhar T. Lower extremity nerve decompression for diabetic peripheral neuropathy: a systematic review and meta-analysis [J]. Plast Reconstr Surg Glob Open, 2022, 10(8): E4478.
[297] Tu Y, Lineaweaver WC, Chen Z, et al. Surgical decompression in the treatment of diabetic peripheral neuropathy: a systematic review and meta-analysis [J]. J Reconstr Microsurg, 2017, 33(3): 151-157.
[298] Macaré van Maurik JF, Oomen RT, van Hal M, et al. The effect of lower extremity nerve decompression on health-related quality of life and perception of pain in patients with painful diabetic polyneuropathy: a prospective randomized trial [J]. Diabet Med, 2015, 32(6): 803-809.
[299] Ang L, Kim HJ, Heo JW, et al. Acupuncture for the treatment of trigeminal neuralgia: a systematic review and meta-analysis [J]. Complement Ther Clin Pract, 2023, 52: 101763.
[300] Yin Z, Wang F, Sun M, et al. Acupuncture methods for primary trigeminal neuralgia: a systematic review and network meta-analysis of randomized controlled trials [J]. Evid Based Complement Alternat Med, 2022, 2022: 3178154.
[301] He HX, Li YX, Xiao YS, et al. The efficacy of acupuncture for trigeminal neuralgia: an overview of systematic reviews [J]. Front Neurol, 2024, 15: 1375587.
[302] Li YX, Li J, Zhang Y, et al. Overview of systematic reviews of acupuncture for trigeminal neuralgia [J]. Liaoning Journal of Traditional Chinese Medicine 2018, 45(11): 2251-2254.
[303] Wang Y, Li W, Peng W, et al. Acupuncture for postherpetic neuralgia: systematic review and meta-analysis [J]. Medicine (Baltimore), 2018, 97(34): E11986.
[304] Pei W, Zeng J, Lu L, et al. Is acupuncture an effective postherpetic neuralgia treatment? A systematic review and meta-analysis [J]. J Pain Res, 2019, 12: 2155-2165.
[305] Huang R, Dong ZW, Yan CC. Systematic assessment and meta-analysis on acupuncture for postherpetic neuralgia [J]. Journal of Traditional Chinese Medical Sciences, 2022, 12(24): 32-36.
[306] Li X, Liu Y, Jing Z, et al. Effects of acupuncture therapy in diabetic neuropathic pain: a systematic review and meta-analysis [J]. Complement Ther Med, 2023, 78: 102992.
[307] Zhou L, Wu T, Zhong Z, et al. Acupuncture for painful diabetic peripheral neuropathy: a systematic review and meta-analysis [J]. Front Neurol, 2023, 14: 1281485.
[308] Zhang X, Xiao L, Qin Y, et al. Acupuncture for the treatment of diabetic peripheral neuropathy in the elderly: a systematic review and meta-analysis [J]. Front Med (Lausanne), 2024, 11: 1339747.
[309] Baviera AF, Olson K, Paula JM, et al. Acupuncture in adults with chemotherapy-induced peripheral neuropathy: a systematic review [J]. Rev Lat Am Enfermagem, 2019, 27: E3126.
[310] Chien TJ, Liu CY, Fang CJ, et al. The efficacy of acupuncture in chemotherapy-induced peripheral neuropathy: systematic review and meta-analysis [J]. Integr Cancer Ther, 2019, 18: 1534735419886662.
[311] Jin Y, Wang Y, Zhang J, et al. Efficacy and safety of acupuncture against chemotherapy-induced peripheral neuropathy: a systematic review and meta-analysis [J]. Evid Based Complement Alternat Med, 2020, 2020: 8875433.
[312] Pei LX, Yi Y, Guo J, et al. The effectiveness and safety of acupuncture/electroacupuncture for chemotherapy-induced peripheral neuropathy: a systematic review and meta-analysis [J]. Acupunct Med, 2023, 41(2): 73-85.
[313] Xu Z, Wang X, Wu Y, et al. The effectiveness and safety of acupuncture for chemotherapy-induced peripheral neuropathy: a systematic review and meta-analysis [J]. Front Neurol, 2022, 13: 963358.
[314] Zhao H, Wang C, Wang X, et al. Efficacy and safety of acupuncture in the treatment of radicular cervical spondylosis: a systematic review and meta-analysis [J]. Comb Chem High Throughput Screen, 2023.
[315] Zhang Z, Hu T, Huang P, et al. The efficacy and safety of acupuncture therapy for sciatica: a systematic review and meta-analysis of randomized controlled trails [J]. Front Neurosci, 2023, 17: 1097830.
[316] 杨世宁, 高翔, 姚勇, 等. 针灸治疗神经根型颈椎病的系统评价及Meta分析[Acupuncture and moxibustion and moxibustion for treatment of cervical spondylotic radiculopathy: A systematic review and meta-analysis] [J]. 甘肃科技, 2021, 37(23): 143-150.
[317] He K, Hu R, Huang Y, et al. Effects of acupuncture on neuropathic pain induced by spinal cord injury: a systematic review and meta-analysis [J]. Evid Based Complement Alternat Med, 2022, 2022: 6297484.
[318] 曾亮, 赵冉, 潘国良, 等. 温针灸治疗神经根型颈椎病随机对照研究的Meta分析 [Effectiveness of Warming Needle Moxibustion for Cervical Apondylotic Radiculopathy:A Meta-analysis] [J]. 按摩与康复医学, 2021, 12(21): 60-64.
[319] 杨伟伟, 王清玉, 李良华. 温针灸治疗神经根型颈椎病疗效 Meta分析[Curative Effect of Warm Needling Moxibustion in Treating Cervical Spondylotic Radiculopathy:A Meta-Analysis] [J]. 福建中医药, 2022, 53(2): 42-47.
[320] 王艳萍, 商洪才, 王飞, 等. 热敏灸治疗带状疱疹后神经痛疗效与安全性的系统评价和Meta分析 [Systematic Evaluation and Meta-analysis on the Therapeutic Effect and Safety of Heat-Sensitive Moxibustion in the Treatment of Postherpetic Neuralgia] [J]. 光明中医, 2021, 36(5): 675-680.
[321] 胡昭端, 周晓红, 谢有琼, 等. 热敏灸治疗神经根型颈椎病的系统评价与meta分析 [Heat-sensitive moxibustion for the treatment ofcervical spondylotic radiculopathy:a systematic review and meta-analysis] [J]. 颈腰痛杂志, 2023, 44(1): 24-28.
[322] 余勇, 马剑桥, 谢悦, 等. 热敏灸治疗神经根型颈椎病疗效的系统评价和Meta分析 [A Systematic Review and Meta-analysis of Therapeutic Effect of Cervical Spondylotic Radiculopathy by Treatment of Heat-sensitive Moxibustion] [J]. 预防医学情报杂志, 2021, 37(9): 1303-1310. (In Chinese)
[323] 吴萍, 徐铭阳, 张媛媛, 等. 电针治疗原发性三叉神经痛的Meta分析[A Meta-analysis of Electroacupuncture Treatment on Primary Trigeminal Neuralgia] [J]. 中国中医急症, 2017, 26(5): 830-833.
[324] 周杰, 梁宜, 陈勤, 等. 电针治疗三叉神经痛随机对照研究的 Meta分析[A Meta-analysis of Randomized Controlled Trials of Aelectroacupuncture Treatment for Trigeminal Neuralgia] [J]. 上海针灸杂志, 2017, 36(4): 478-483.
[325] 刘盈君, 张全爱, 吴媛媛, 等. 电针治疗带状疱疹后遗神经痛临床疗效及安全性的Meta分析[Meta-analysis for Efficacy and Safety of Electroacupuncture in Treating Postherpetic Neuralgia] [J]. 广州中医药大学学报, 2020, 37(12): 2472-2480.
[326] Zhang T, Zhang Q, Zhu P, et al. The efficacy of acupuncture in the treatment of chemotherapy-induced peripheral neuropathy: a network meta-analysis [J]. Altern Ther Health Med, 2023, 29(8): 898-906. PMID: 37708563.
[327] 邝碧瑶, 李敏, 曾欢, 等. 电针治疗化疗所致周围神经病变的Meta分析[Meta-analysis of electroacupuncture for chemotherapy-induced peripheral neuropathy] [J]. 天津中医药, 2019, 36(7): 673-678.
[328] 粟胜勇, 李妮娜, 赵骏, 等. 电针治疗神经根型颈椎病临床疗效Meta分析[Clinical Effect of Electrol-acupuncture Treatment of Cervical Spondylotic Radiculopathy : A Meta Analysis] [J]. 辽宁中医药大学学报, 2017, 19(8): 9-11.
[329] 高浚洋, 卢春键, 袁金筠, 等. 火针治疗带状疱疹后遗神经痛疗效及安全性的Meta分析[The Efficacy and Safety of Fire Needling in the Treatment of Postherpetic Neuralgia:A Meta-analysis] [J]. 广州中医药大学学报, 2023, 40(9): 2403-2410.
[330] 黄守强, 熊俊, 项洁, 等. 火针治疗带状疱疹后遗神经痛有效性和安全性的系统评价[Efficacy and safety of fire needle for post-herpetic neuralgia:a systematic review] [J]. 中国循证医学杂志, 2022, 22(2): 168-175.
[331] 唐兴, 陈星良,徐定涛, 等. 火针疗法治疗带状疱疹后遗神经痛疗效的Meta分析[Meta Analysis of the Efficacy of Fire Needle Therapy in the Treatment of Postherpetic Neuralgia] [J]. 世界最新医学信息文摘(连续型电子期刊), 2021, 21(12): 268-269, 271.
[332] 洪婉仪, 江钢辉, 刘冰清. 浮针治疗带状疱疹后遗神经痛随机对照试验的Meta分析和试验序贯分析 [Meta-analysis and Trial Sequential Analysis of Randomized Controlled Trials of Fu's Subcutaneous Needling for Postherpetic Neuralgia] [J]. 广州中医药大学学报, 2023, 40(1): 261-270.
[333] Zhang B, Yin LW, Zhu HH, et al. Meta-analysis of Fu's Subcutaneous Needling in the Treatment of Postherpetic Neuralgia [J]. Journal of Guangzhou University of Chinese Medicine, 2023, 40(8): 2108-2117.
[334] Chen SY, Li XJ, Liu YW, et al. Meta-analysis of efficacy and safety of treating post herpetic neuralgia by percussopunctator [J]. Clinical Journal of Chinese Medicine, 2023, 15(35): 73-78.
[335] Peng Y, Wu J, Zhang T, et al. Needle-knife therapy for post herpetic neuralgia: a systematic review and meta-analysis [J]. Asian J Surg, 2024, 47(1): 704-706.
[336] Zhao MM, Liu FS, Hong T, et al. Systematic review of acupotomy for cervical spondylotic radiculopathy [J]. Traditional Chinese Medicine Journal, 2016, 15(4): 40-42, 45. (In Chinese)
[337] Tan Y, Hu J, Pang B, et al. Moxibustion for the treatment of diabetic peripheral neuropathy: a systematic review and meta-analysis following PRISMA guidelines [J]. Medicine (Baltimore), 2020, 99(39): E22286.
[338] Zhou, DN, Qi, FJ, Wang, ZJ, et al.. Meta-analysis of Mainly Treated with Pricking Blood and Cupping in the Treatment of Postherpetic Neuralgia [J]. Journal of Liaoning University of Traditional Chinese Medicine, 2022, 24(8): 166-171.
[339] Li CL, Ma J. Meta-analysis of Acupoint Injection fRR Postherpetic Neuralgia [J]. World Latest Medicine Information (Electronic Version), 2020, 20(A2): 37 41.
[340] Lin XF, Tao K, Zhang GL, et al. Systematic review of the therapeutic effect of Jiaji points injection on postherpetic neuralgia caused by herpes zoster [J]. Journal of Modern Medicine & Health, 2023, 39(21): 3685 3690.
[341] Pu B, Lu X, Yu P, et al. Acupoint herbal patching for postherpetic neuralgia: a systematic review and meta-analysis [J]. Medicine (Baltimore), 2024, 103(4): E37029.
[342] Xie B, Wang Q, Zhou C, et al. Efficacy and safety of the injection of the traditional chinese medicine puerarin for the treatment of diabetic peripheral neuropathy: a systematic review and meta-analysis of 53 randomized controlled trials [J]. Evid Based Complement Alternat Med, 2018, 2018: 2834650.
[343] Wu J, Zhang X, Zhang B. Efficacy and safety of puerarin injection in treatment of diabetic peripheral neuropathy: a systematic review and meta-analysis of randomized controlled trials [J]. J Tradit Chin Med, 2014, 34(4): 401-10.
[344] Chen CY, Chen PH, Chen B, et al. Systematic Evaluation of Huangqi Guizhi Wuwutang in the Treatment of Cervical Spondylotic Radiculopathy [J]. Rheumatism and Arthritis, 2018, 7(2): 37-42.
[345] Fu ZW, Qu LX. Meta-Analysis of Huangqi Guizhi Wuwu Decoction for Treating Cervical Radiculopathy [J]. Modern Medical Journal, 2022, 50(4): 434-440.
[346] Yu X, Li XH. Meta-analysis of Clinical Efficacy of Xuefu Zhuyu Decoction in the Treatment of PHN [J]. Smart Healthcare, 2022, 8(34): 75-78, 82.
[347] Zhao E, Wang J, Li Q, et al. Efficacy of Xuefu-Zhuyu decoction in treatment of postherpetic neuralgia: a Meta-analysis [J]. Journal of Xinjiang Medical University, 2023, 46(3): 390-397.
[348] Meizhen Z, Xiaohui H, Yiting T, et al. Efficacy and safety of Buyang Huanwu decoction for diabetic peripheral neuropathy: a systematic review and metaanalysis [J]. J Tradit Chin Med, 2023, 43(5): 841-850.
[349] Wang YM, Leng JH. Meta analysis of the clinical efficacy of Shentong Zhuyu Decoction in the treatment of diabetic peripheral neuropathy [J]. Journal of China Prescription Drug, 2024, 22(2): 41-45.
[350] Han J, Duan XY. Meta-analysis of Shaoyao Gancao Decoction in the Treatment of Postherpetic Neuralgia [J]. Journal of Kunming University, 2023, 45(6): 111-116.
[351] Scott W, Guildford BJ, Badenoch J, et al. Feasibility randomized-controlled trial of online acceptance and commitment therapy for painful peripheral neuropathy in people living with HIV: the OPEN study [J]. Eur J Pain, 2021, 25(7): 1493-1507.
[352] Bäckryd E, Ghafouri N, Gerdle B, et al. Rehabilitation interventions for neuropathic pain: a systematic review and meta-analysis of randomized controlled trials [J]. J Rehabil Med, 2024, 56: jrm40188.
[353] Langlois P, Perrochon A, David R, et al. Hypnosis to manage musculoskeletal and neuropathic chronic pain: a systematic review and meta-analysis [J]. Neurosci Biobehav Rev, 2022, 135: 104591.
[354] McKittrick ML, Connors EL, McKernan LC. Hypnosis for chronic neuropathic pain: a scoping review [J]. Pain Med, 2022, 23(5): 1015-1026.
[355] Higgins DM, Heapy AA, Buta E, et al. A randomized controlled trial of cognitive behavioral therapy compared with diabetes education for diabetic peripheral neuropathic pain [J]. J Health Psychol, 2022, 27(3): 649-662.
[356] Bai Y, Ma JH, Yu Y, et al. Effect of cognitive-behavioral therapy or mindfulness therapy on pain and quality of life in patients with diabetic neuropathy: a systematic review and meta-analysis [J]. Pain Manag Nurs, 2022, 23(6): 861-870.
[357] Meize-Grochowski R, Shuster G, Boursaw B, et al. Mindfulness meditation in older adults with postherpetic neuralgia: a randomized controlled pilot study [J]. Geriatr Nurs, 2015, 36(2): 154-160.
[358] Izgu N, Gok Metin Z, Karadas C, et al. Progressive muscle relaxation and mindfulness meditation on neuropathic pain, fatigue, and quality of life in patients with type 2 diabetes: a randomized clinical trial [J]. J Nurs Scholarsh, 2020, 52(5): 476-487.
[359] Hussain N, Said ASA. Mindfulness-based meditation versus progressive relaxation meditation: impact on chronic pain in older female patients with diabetic neuropathy [J]. J Evid Based Integr Med, 2019, 24: 2515690X19876599.
[360] Johannsen M, O'Connor M, O'Toole MS, et al. Efficacy of mindfulness-based cognitive therapy on late post-treatment pain in women treated for primary breast cancer: a randomized controlled trial [J]. J Clin Oncol, 2016, 34(28): 3390-3399.
[361] Rozworska KA, Poulin PA, Carson A, et al. Mediators and moderators of change in mindfulness-based stress reduction for painful diabetic peripheral neuropathy [J]. J Behav Med, 2020, 43(2): 297-307.
[362] Thuma K, Ditsataporncharoen T, Arunpongpaisal S, et al. Hypnosis as an adjunct for managing pain in head and neck cancer patients post radiotherapy [J]. J Med Assoc Thai, 2016, 99(Suppl 5): S141-S147. PMID: 29906024.
[363] Rivaz M, Rahpeima M, Khademian Z, et al. The effects of aromatherapy massage with lavender essential oil on neuropathic pain and quality of life in diabetic patients: a randomized clinical trial [J]. Complement Ther Clin Pract, 2021, 44: 101430.
[364] Gok Metin Z, Arikan Donmez A, Izgu N, et al. Aromatherapy massage for neuropathic pain and quality of life in diabetic patients [J]. J Nurs Scholarsh, 2017, 49(4): 379-388.
Cite This Article
  • APA Style

    Zhixiang, C., Yan, L., Xianguo, L., Lingjie, X., Dong, H., et al. (2025). A Chinese Guideline for the Diagnosis and Treatment of Neuropathic Pain. International Journal of Pain Research, 1(4), 56-86. https://doi.org/10.11648/j.ijpr.20250104.12

    Copy | Download

    ACS Style

    Zhixiang, C.; Yan, L.; Xianguo, L.; Lingjie, X.; Dong, H., et al. A Chinese Guideline for the Diagnosis and Treatment of Neuropathic Pain. . 2025, 1(4), 56-86. doi: 10.11648/j.ijpr.20250104.12

    Copy | Download

    AMA Style

    Zhixiang C, Yan L, Xianguo L, Lingjie X, Dong H, et al. A Chinese Guideline for the Diagnosis and Treatment of Neuropathic Pain. . 2025;1(4):56-86. doi: 10.11648/j.ijpr.20250104.12

    Copy | Download

  • @article{10.11648/j.ijpr.20250104.12,
      author = {Cheng Zhixiang and Lu Yan and Liu Xianguo and Xia Lingjie and Huang Dong and Song Tao and Liang Lishuang and Zhang Ying and Liu Tanghua and Wang Lin and Tao Wei and Zhang Baojuan and Zhang Xiaomei and Feng Zeguo and Wang Jianxiu and Wang Suoliang and Liu Yanqing},
      title = {A Chinese Guideline for the Diagnosis and Treatment of Neuropathic Pain
    },
      journal = {International Journal of Pain Research},
      volume = {1},
      number = {4},
      pages = {56-86},
      doi = {10.11648/j.ijpr.20250104.12},
      url = {https://doi.org/10.11648/j.ijpr.20250104.12},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.ijpr.20250104.12},
      abstract = {Background: Neuropathic pain refers to pain caused by injury or disease affecting the somatosensory nervous system, which is a common and frequently occurring disease in clinical practice, and seriously affects patients' quality of life. However, the treatment of neuropathic pain is a clinical challenge. Objective: To standardize neuropathic pain management, the Pain Medicine Branch of China Association of Health Care for the Elderly convened an expert panel to develop the guideline. Main ideas: Based on high quality evidence-based medical research on the diagnosis and treatment of neuropathic pain published domestically and internationally in the past 10 years, the expert group has formed recommendations for common treatment methods through rigorous argumentation and expert voting, to provide references for standardized diagnosis and treatment of neuropathic pain. This guideline adopts GRADE methodology to evaluate the level of evidence and strength of recommendation for the treatments of common peripheral neuropathic pain and central neuropathic pain. Chinese traditional medicine also plays an important role in the treatment of neuropathic pain, so this guide also provides Chinese traditional medicine drugs and treatment recommendations. Conclusion: The Chinese pain community has proposed the principle of "treatment forward, early intervention, prevention of sensitization, and prevention and treatment of chronic pain", which has played a positive role in improving the clinical diagnosis and treatment level of neuropathic pain.},
     year = {2025}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - A Chinese Guideline for the Diagnosis and Treatment of Neuropathic Pain
    
    AU  - Cheng Zhixiang
    AU  - Lu Yan
    AU  - Liu Xianguo
    AU  - Xia Lingjie
    AU  - Huang Dong
    AU  - Song Tao
    AU  - Liang Lishuang
    AU  - Zhang Ying
    AU  - Liu Tanghua
    AU  - Wang Lin
    AU  - Tao Wei
    AU  - Zhang Baojuan
    AU  - Zhang Xiaomei
    AU  - Feng Zeguo
    AU  - Wang Jianxiu
    AU  - Wang Suoliang
    AU  - Liu Yanqing
    Y1  - 2025/10/14
    PY  - 2025
    N1  - https://doi.org/10.11648/j.ijpr.20250104.12
    DO  - 10.11648/j.ijpr.20250104.12
    T2  - International Journal of Pain Research
    JF  - International Journal of Pain Research
    JO  - International Journal of Pain Research
    SP  - 56
    EP  - 86
    PB  - Science Publishing Group
    UR  - https://doi.org/10.11648/j.ijpr.20250104.12
    AB  - Background: Neuropathic pain refers to pain caused by injury or disease affecting the somatosensory nervous system, which is a common and frequently occurring disease in clinical practice, and seriously affects patients' quality of life. However, the treatment of neuropathic pain is a clinical challenge. Objective: To standardize neuropathic pain management, the Pain Medicine Branch of China Association of Health Care for the Elderly convened an expert panel to develop the guideline. Main ideas: Based on high quality evidence-based medical research on the diagnosis and treatment of neuropathic pain published domestically and internationally in the past 10 years, the expert group has formed recommendations for common treatment methods through rigorous argumentation and expert voting, to provide references for standardized diagnosis and treatment of neuropathic pain. This guideline adopts GRADE methodology to evaluate the level of evidence and strength of recommendation for the treatments of common peripheral neuropathic pain and central neuropathic pain. Chinese traditional medicine also plays an important role in the treatment of neuropathic pain, so this guide also provides Chinese traditional medicine drugs and treatment recommendations. Conclusion: The Chinese pain community has proposed the principle of "treatment forward, early intervention, prevention of sensitization, and prevention and treatment of chronic pain", which has played a positive role in improving the clinical diagnosis and treatment level of neuropathic pain.
    VL  - 1
    IS  - 4
    ER  - 

    Copy | Download

Author Information