Skip to main content

Multimodal non-invasive non-pharmacological therapies for chronic pain: mechanisms and progress

Abstract

Background

Chronic pain conditions impose significant burdens worldwide. Pharmacological treatments like opioids have limitations. Non-invasive non-pharmacological therapies (NINPT) encompass diverse interventions including physical, psychological, complementary and alternative approaches, and other innovative techniques that provide analgesic options for chronic pain without medications.

Main body

This review elucidates the mechanisms of major NINPT modalities and synthesizes evidence for their clinical potential across chronic pain populations. NINPT leverages peripheral, spinal, and supraspinal mechanisms to restore normal pain processing and limit central sensitization. However, heterogeneity in treatment protocols and individual responses warrants optimization through precision medicine approaches.

Conclusion

Future adoption of NINPT requires addressing limitations in standardization and accessibility as well as synergistic combination with emerging therapies. Overall, this review highlights the promise of NINPT as a valuable complementary option ready for integration into contemporary pain medicine paradigms to improve patient care and outcomes.

Peer Review reports

Background

Chronic pain conditions, such as osteoarthritis, diabetic neuropathy, and fibromyalgia, refer to a range of complex and multifaceted disorders that affect millions of individuals worldwide and impose significant burdens on society, healthcare systems, and personal well-being [1, 2]. The management of chronic pain presents a major challenge for healthcare professionals and researchers, as there is no single or universally effective treatment and patients often suffer from chronic pain for a long period of time [3, 4]. Furthermore, conventional treatments such as opioids and nonsteroidal anti-inflammatory drugs (NSAIDs) carry significant risks and limitations, including addiction, tolerance, dependence, gastrointestinal bleeding, renal impairment, and cardiovascular events, especially when used long term [5, 6]. As a result, additional approaches are needed to provide safe and effective chronic pain relief without causing harm or adverse effects [7].

Non-invasive non-pharmacological therapies (NINPT) offer options for managing chronic pain safely and effectively without damaging tissues or causing significant side effects [8]. For chronic pain conditions, NINPT can be categorized into physical modalities, psychological interventions, complementary and alternative therapies, and other innovative techniques [8, 9]. Physical modalities like transcutaneous electrical nerve stimulation (TENS), heat therapy and cryotherapy, and massage aim to modulate chronic pain signals through physiological mechanisms [10,11,12]. Psychological interventions including cognitive-behavioral therapy (CBT) and mindfulness-based therapy help patients cope with chronic pain by promoting positive psychological and behavioral changes [13,14,15]. Complementary and alternative therapies such as acupuncture, yoga, and music therapy provide additional approaches to reducing chronic pain through mechanisms involving both physical and psychological aspects [16,17,18]. Other interventions like chiropractic care, biofeedback, and virtual reality (VR) therapy employ techniques to modulate chronic pain [19,20,21]. For chronic pain management, NINPT can be used alone or combined with conventional treatments based on specific conditions, patient preferences, and availability of resources.

Furthermore, understanding the mechanisms underlying how NINPT alleviate chronic pain is critical for optimizing their clinical application. To begin with, it helps establish the causal relationship between NINPT and their effects on chronic pain, clarifying their validity and reliability [15, 22]. Secondly, elucidating the mechanisms of NINPT action allows for optimized, individualized therapeutic regimens meeting the needs of each patient. This includes determining the optimal frequency, intensity, and duration for achieving the best outcomes [23]. Thirdly, it spurs continued innovation in developing more effective and integrative NINPT modalities for chronic pain relief through emerging technologies [7, 24]. Lastly, establishing the neurophysiological mechanisms underlying the effects of NINPT on chronic pain enables the development of evidence-based clinical guidelines on recommending appropriate NINPT options for different chronic pain conditions [9, 25].

The objective of this review is to provide a comprehensive and up-to-date overview of various NINPT for chronic pain management, with a focus on their mechanisms of action and clinical applications. For each modality, we discuss how they modulate pain signaling across peripheral, spinal, and supraspinal levels. We also highlight limitations, challenges, and future directions for optimizing the use of NINPT. We aim to build a bridge between basic research on mechanisms of NINPT and their clinical implementation for chronic pain relief.

Theoretical backgrounds of chronic pain

While acute and chronic pain both involve the pain pathway from nociceptors to brain, chronic pain arises from dysregulation of the balanced mechanisms that adaptively modulate pain signaling in the acute state. Elucidating the neurophysiological processes underlying this maladaptive transition across molecular, cellular, and systems levels will inform targeted interventions.

Anatomy and physiology of pain pathways

The pain pathway consists of three core components: nociceptors that detect pain signals, the spinal cord that transmits and modulates the signals, and the brain that processes and regulates the pain experience [26]. Nociceptors are specialized sensory receptors that respond to harmful stimuli by converting them into electrical signals. In chronic pain, nociceptors become hypersensitive and can be activated by normal sensory stimulation. These signals are transmitted by Aδ and C fibers to the dorsal horn of the spinal cord, where they synapse with second-order neurons [27] (Fig. 1).

Fig. 1
figure 1

Ascending pain pathway from periphery to central. Note: PAG = periaqueductal gray

The dorsal horn of the spinal cord is the first site where pain signals enter the spinal cord and undergo initial processing and modulation [26]. In chronic pain, the dorsal horn shows increased neuronal excitability and reduced inhibition, amplifying and dysregulating pain signal transmission. Neurons and neurotransmitters in the dorsal horn, like glutamatergic and GABAergic neurons, facilitate and inhibit pain signal conduction. Descending facilitation from the brainstem also enhances the dorsal horn’s role in augmenting pain signaling in chronic pain [27].

The second-order neurons then cross the midline or remain ipsilateral and ascend to the brain through tracts including the spinothalamic, spinoreticular, and spinomesencephalic tracts [26] (Fig. 1). In chronic pain, changes in the volume and connectivity of these tracts correlate with clinical pain intensity and modulation. Pain signals are processed and modulated in various brain regions, including the thalamus, brainstem, and cortex. Multiple brain areas exhibit functional and anatomical changes in chronic pain that disrupt normal pain modulation [28, 29].

Recent research has advanced our understanding of the mechanisms underlying pain signaling and modulation across the pain pathway. Targeting these maladaptive changes may help restore normal processing, facilitate adaptation and improve quality of life. At the nociceptor level, factors like cytokines, growth factors, and nerve injury regulate ion channels including sodium, potassium, calcium, and transient receptor potential channels [30, 31]. Neurotransmitter interactions in the spinal cord, such as glutamate, GABA, glycine, and substance P, play an essential role in synaptic transmission and plasticity of pain signals [32, 33] (Fig. 2).

Fig. 2
figure 2

Modulation of pain signals in the spinal dorsal horn

Various brain regions, including the anterior cingulate cortex, insula, amygdala, prefrontal cortex, and periaqueductal gray, detect, process, and modulate pain signals by activating or inhibiting circuits involved in pain’s sensory, emotional, cognitive, and behavioral aspects [29, 34, 35]. The endogenous opioid system also regulates pain modulation through inhibiting signaling and enhancing analgesia. Changes in these modulatory systems underpin disrupted pain regulation in chronic pain (Fig. 3).

Fig. 3
figure 3

Interactions between pain modulatory brain regions. Note: The red lines represent the ascending nociceptive pathways that transmit pain signals, while the blue lines represent the descending anti-nociceptive pathways that modulate pain. PFC = prefrontal cortex; ACC = anterior cingulate cortex; IC = insula cortex; PAG = periaqueductal gray; RVM = rostral ventromedial medulla

Disruption of pain modulation

Pain modulation refers to the process by which nociceptive signals are regulated to achieve balanced sensitivity to potential threats. In acute pain, inhibition dominates over facilitation, keeping modulation intact. In chronic pain, modulation becomes impaired due to disrupted inhibition and enhanced facilitation across the pain pathway.

In the dorsal horn, normal modulation involves balanced excitatory and inhibitory controls by neurotransmitters like glutamate, GABA, and glycine [33]. Descending facilitation and inhibition from the brainstem also contribute to regulating pain sensitivity. In chronic pain, increased descending facilitation, impaired GABA/glycine inhibition, and neuroinflammation enhance dorsal horn neuron excitability, disrupting modulation. Prolonged nociceptive signaling leads to central sensitization, further amplifying pain signaling [36].

In the brain, the anterior cingulate cortex and periaqueductal gray are critical for modulation [37]. The anterior cingulate cortex exerts cognitive and emotional modulation, while periaqueductal gray connections provide downstream inhibitory control [38]. In chronic pain, reduced connectivity between these areas undermines enhanced pain inhibition. The prefrontal cortex also shows impaired cognitive modulation [29, 39]. Changes in endogenous modulatory systems, including opioids, further weaken inhibitory control.

Various interventions aim to rebalance modulation across the pain pathway. Targeting abnormal changes at all neuromatrix levels may help reinstate neural balance, regain function, and improve quality of life. A mechanism-based precision approach may address individual heterogeneity in chronic pain [40].

In summary, the progression from acute to chronic pain signifies a transition from an adaptable nociceptive system with modulated pain sensitivity to one dominated by unregulated signaling across the nervous system. Restoring modulation is key to limiting chronicity and optimizing outcomes. Overall, chronic pain arises from disruption of balanced mechanisms that regulate nociceptive signal perception and transmission in the normal acute pain state.

Central sensitization: a driver of chronic pain

Central sensitization refers to the maladaptive upregulation of the central nervous system’s response to painful stimuli and normal sensory signals. Through descending facilitation, widespread hyperexcitability, impaired inhibition, and inflammatory processes, central sensitization manifests as an amplification of neural signaling along the neuraxis that drives chronic pain [41, 42]. It represents a key intermediate mechanism between acute and chronic pain. Neuroinflammation also plays an important role in the development and maintenance of central sensitization.

At the spinal level, constant nociceptive input from peripheral pain generators drives hyperexcitability of dorsal horn neurons, known as heterosynaptic facilitation [43]. Glutamate and inflammatory mediators activate NMDA receptors on dorsal horn neurons, initiating events that enhance neuronal excitability and synaptic plasticity [44]. Loss of inhibitory control mediated by neurotransmitters like GABA and glycine further amplifies signaling to higher brain centers. Descending facilitation from the brainstem exacerbates spinal hyperexcitability [44].

In the brain, key structures involved in the sensory, emotional and cognitive aspects of pain show altered activity, connectivity, and neurochemistry. For example, the anterior cingulate cortex exhibits elevated activity but reduced connectivity with the periaqueductal gray, diminishing engagement of descending inhibitory controls [45, 46]. The prefrontal cortex shows impaired cognitive control over pain [39]. Changes in the endogenous opioid system and microglial activation highlight the role of disrupted top-down modulation and neuroinflammation in perpetuating central sensitization.

Processes like long-term potentiation, epigenetic changes, and neuroimmune dysregulation have been implicated in the cellular and molecular processes underlying central sensitization. Targeting the mechanisms driving and maintaining central sensitization may help restore normal pain regulation, limit chronicity, and improve wellbeing. Treatment needs a systematic approach addressing changes across the pain neuromatrix and balancing neural modulation. Central sensitization represents a seminal mechanism in pain chronification demanding mechanism-based management [47]. Overall, central sensitization signifies an enduring state of maladaptive neuroplasticity across the neuraxis that amplifies pain transmission and disables endogenous modulation.

In summary, central sensitization promotes chronic pain by inducing a self-perpetuating cycle of enhanced signaling and diminished modulation across the pain neuraxis.

Types of NINPT

NINPT encompass heterogeneous physical, psychological, and complementary therapies as well as other techniques that modulate multidimensional aspects of the pain pathway without invasive procedures. Understanding their differential effects and mechanisms of action at peripheral, spinal, and supraspinal sites will enable optimal translation and integration of these modalities to relieve chronic pain.

Physical modalities

Physical modalities are interventions that use physical stimuli, such as electricity, heat, cold, or pressure, to modulate pain signals. Physical modalities can act on the peripheral, spinal, or supraspinal levels of the pain pathway, depending on the type, intensity, duration, and location of the stimuli. Some of the most common physical modalities are:

TENS

TENS is a method for relieving pain through the application of low-intensity electrical stimulation. However, repeated TENS applications may cause analgesic tolerance [48]. This technique is delivered via TENS devices which provide electrical stimulation through leads attached to adhesive electrodes. TENS devices include compact, battery-powered portable units as well as larger stationary models [49, 50].

At the peripheral level, TENS activates large myelinated Aβ fibers which inhibit nociceptive transmission in small Aδ and C fibers according to the gate control theory. This relies on presynaptic inhibition of nociceptive input at the dorsal horn [51, 52]. Molecular studies show TENS modulates the expression and activity of key pain-related ion channels on peripheral nerves, including voltage-gated sodium channels, TRP channels, inhibiting excitability and neurotransmission in nociceptor [53, 54].

In the spinal cord, functional magnetic resonance imaging (MRI) indicates TENS suppresses activity in Rexed laminae I-V of the dorsal horn, which transmit and modulate nociceptive signaling [55, 56]. Immunohistochemistry studies demonstrate TENS increases spinal expression of μ-opioid, δ-opioid, and cannabinoid receptors [57]. Electrophysiology studies show TENS reduces wind-up and potentiation of dorsal horn neurons by modulating NMDA receptors [58]. Repeated use may cause tolerance due to NMDA receptor desensitization [48].

In the brain, TENS alters activity and functional connectivity in regions including the thalamus, somatosensory cortex, anterior cingulate cortex, insula, prefrontal cortex, and periaqueductal gray, suggesting effects on sensory, cognitive, and affective dimensions of pain. Functional connectivity analysis reveals TENS strengthens top-down connectivity from the periaqueductal gray and prefrontal cortex to the anterior cingulate cortex [59]. Electroencephalogram (EEG) shows TENS increases alpha and beta oscillations that correlate with analgesia [60]. Molecular studies indicate TENS increases endogenous opioid and serotonin levels in brain regions involved in descending modulation [61].

Evidence shows TENS can reduce pro-inflammatory cytokines and modulate neuroimmune interactions involved in chronic pain [62]. It may also promote neural plasticity by stimulating neurotrophic factors like brain-derived neurotrophic factor (BDNF) and beta-endorphins [63].

Recent clinical trials support the efficacy of TENS for relieving neuropathic pain in conditions such as diabetic neuropathy, post-herpetic neuralgia, and neuropathic pain after spinal cord injury [64,65,66] (Additional file 1: Fig. S1). Randomized controlled trials demonstrate TENS reduces neuropathic pain intensity and improves physical function compared to sham TENS. The effects may involve altering peripheral nerve excitability and central sensitization [66, 67].

For osteoarthritis pain, meta-analyses of numerous randomized controlled trials indicate TENS applied to the knee joint decreases pain and improves physical function [68, 69]. Proposed mechanisms include reducing inflammation, activating descending inhibition, and stimulating analgesic neurotransmitter release. However, the evidence for TENS in relieving stiffness in osteoarthritis is inconsistent [69, 70]. Although some studies suggest TENS may reduce stiffness in osteoarthritis patients [69], the efficacy of TENS on osteoarthritis needs further verification.

In fibromyalgia, TENS applied to tender points provides analgesia and improves pain threshold compared to sham treatment [71]. Suggested mechanisms involve modulating substance P levels, increasing blood flow, and restoring abnormal pain processing.

While research has focused on the effects of TENS for osteoarthritis and fibromyalgia, future studies should evaluate its therapeutic potential for other common causes of chronic pain such as diabetic neuropathy.

TENS may have synergistic benefits when combined with pharmacological, psychological, physical, or complementary interventions as part of a multimodal approach. For example, TENS plus opioid therapy could improve analgesia while reducing opioid-related risks [72]. TENS combined with CBT may provide sensory-cognitive modulation of chronic pain [73]. Future research should explore optimal multimodal protocols incorporating TENS for diverse chronic pain conditions.

While TENS shows promise, larger, high-quality randomized controlled trials are needed to establish optimal protocols and long-term effectiveness for chronic pain relief. Emerging research on computational modeling and artificial intelligence provides new opportunities to optimize TENS stimulus parameters such as intensity, frequency, and pattern [74]. Advanced algorithms and neural networks could analyze individual patient factors to determine the optimal dosing strategy. Real-time closed-loop TENS systems with biofeedback may also enhance outcomes [75].

Heat therapy and cryotherapy

Heat therapy and cryotherapy refer to the application of thermal agents to tissue for pain relief. Heat therapy utilizes sources such as hot packs, paraffin wax, and infrared radiation to apply superficial or deep heating [76]. Cryotherapy employs cold agents such as ice packs, cold compresses, and refrigerant sprays to lower tissue temperature [76].

Heat therapy enhances blood flow and relaxes muscles, while activating thermosensitive TRP channels that modulate peripheral and central thermal sensitivity [77]. Neuroimaging studies show heat may activate endogenous opioid and cortical serotonergic systems involved in analgesia by improving connectivity between the thalamus, anterior cingulate cortex, and periaqueductal gray [76, 78]. Recent evidence suggests heat therapy reduces central sensitization markers including neuroinflammation and NMDA receptor activation [79, 80].

In contrast, cryotherapy limits inflammation and nerve conduction velocity through vasoconstriction and by suppressing release of neurotransmitters including serotonin and glutamate. This inhibits peripheral sensitization and nociceptive signals transmission [81]. Cryotherapy may also mitigate central sensitization by reducing membrane excitability, presynaptic transmitter release, and signal propagation in pain pathways. EEG studies reveal cryotherapy enhances alpha and beta wave rhythms, which may impact cognitive and attentional pain modulation [82, 83].

Emerging studies propose that heat and cold may also influence neural plasticity and central sensitization in chronic pain. Heat therapy appears to enhance neuroplasticity markers like BDNF and reduces central sensitization markers like NMDA receptor expression [79]. In contrast, cryotherapy may limit central sensitization by reducing neurotransmitter release, metabolism, and nerve conduction velocities [81].

Recent clinical trials demonstrate heat therapy reduces neuropathic pain in conditions like diabetic neuropathy, chemotherapy-induced peripheral neuropathy, and neuropathic pain after spinal cord injury [84,85,86] (Additional file 1: Fig. S1). The effects may involve modifying thermal hyperalgesia and central sensitization. Meta-analyses also indicate cryotherapy decreases neuropathic and nerve injury pain.

For osteoarthritis pain, heat and cold therapies applied to the affected joints provide analgesia and improve physical function versus placebo [87, 88]. The mechanisms may involve altering inflammation, muscle relaxation, and joint stiffness.

In fibromyalgia, localized heat and cryotherapy on tender points alleviates widespread pain and sensitivity compared to control conditions [89]. Possible effects include releasing muscle trigger points and restoring descending modulation.

In addition to osteoarthritis and fibromyalgia, future research should assess the effects of thermal therapies on other frequent causes of chronic pain like nerve injury and central sensitization.

Thermal therapies as adjuvants to pharmacologic treatment like NSAIDs, opioids, and membrane stabilizers demonstrate enhanced analgesia in chronic postoperative pain and neuropathic pain [90, 91]. Heat and cold may exert synergistic effects through sensory-discriminative pathways when combined with analgesics.

Integrating thermal therapies into exercise and physical therapy improves outcomes for chronic musculoskeletal pain compared to either treatment alone. The combination optimizes physiological and psychosocial factors influencing chronic pain [92, 93].

While thermal therapies demonstrate short-term analgesia, long-term effectiveness and optimal protocols require further research through high-quality, large-scale randomized controlled trials with extended follow-up periods [94]. There is a lack of evidence on the duration of pain relief and ideal treatment frequency and dosing [92]. Future studies should investigate long-term impacts on pain reduction, functional improvement, side effects, and quality of life.

Individualization of thermal therapies is essential to maximize analgesic effects while ensuring safety and tolerability. Treatment plans should be tailored based on pain location and type, patient preferences, and degree of desired temperature change [92, 95]. Factors like comorbidities, age, sex, and individual variation in thermal sensitivity and endogenous analgesia systems may influence outcomes. Real-time monitoring and feedback could help determine optimal personalized protocols.

Proper safety precautions should be taken during delivery of heat and cold to avoid adverse events like burns, frostbite, or nerve damage. Heat therapy risks include dehydration and aggravated inflammation if overheated [96]. Prolonged exposure to extreme cold may cause irreversible nerve injury [94]. Application guidelines regarding duration, temperature thresholds, and monitoring should be followed, especially in special populations. Overall, individualization of thermal therapies while ensuring safety is key to optimizing effectiveness.

Massage therapy

Massage therapy has emerged as an effective method for managing chronic pain with promising results in clinical trials [12]. However, the underlying mechanisms by which massage relieves chronic pain remain unclear.

The gate control theory suggests massage activates spinal inhibitory interneurons to suppress pain signal transmission to the central nervous system. Massage may activate cutaneous mechanoreceptors and modulate spinal processing of tactile inputs [97]. Neuroimaging studies reveal massage increases release of endogenous opioids like beta-endorphins, enhancing descending modulation from the periaqueductal gray [98]. Massage regulates autonomic balance by increasing parasympathetic activity and reducing inflammatory cytokines [12].

Massage could modulate the hypothalamic–pituitary–adrenal axis and sympathetic nervous system involved in regulating inflammation and stress responses. This may decrease pro-inflammatory cytokines like Interleukin (IL)-6 and tumor necrosis factor alpha (TNF-α) while increasing anti-inflammatory cytokines like IL-10 [12]. Downregulation of pro-inflammatory cytokines may reduce sensitization of peripheral nociceptors and central pain pathways.

Recent evidence indicates massage therapy could prevent or reverse central sensitization by normalizing aberrant sensory input, improving circulation, decreasing trigger points and muscle tension, and optimizing descending inhibition. Brain stimulation studies suggest massage normalizes functional connectivity in chronic pain-related neural networks including the default mode, salience and sensorimotor systems [99].

Recent randomized controlled trials demonstrate massage therapy decreases neuropathic pain in diabetic neuropathy, chemotherapy-induced peripheral neuropathy, and nerve injury conditions [100,101,102] (Additional file 1: Fig. S1). The effects may involve modulating neural hypersensitivity and plasticity.

For osteoarthritis pain, massage applied to affected joints consistently reduces pain and improves physical function compared to control interventions. Massage alleviates pain by enhancing joint mobility, muscle relaxation, and circulation [103].

In migraine and tension headaches, massage therapy targeting neck, head, and facial muscles significantly reduces headache frequency, intensity, and disability versus placebo [104]. The mechanisms likely involve inhibiting peripheral and central sensitization.

As an adjuvant, massage combined with pharmacotherapy like NSAIDs, tricyclics, and anticonvulsants shows greater analgesia than drugs alone in chronic back pain and fibromyalgia [105, 106]. Massage may enhance drug absorption and act synergistically on pain pathways.

For musculoskeletal conditions like osteoarthritis, neck pain, and shoulder pain, massage integrated into exercise and physical therapy improves outcomes including pain relief, function, and quality of life compared to either treatment alone [12, 107].

While massage demonstrates short-term pain relief, evaluating its long-term efficacy requires assessing multidimensional outcomes including sustained pain reduction, functional improvement in daily activities, quality of life enhancement, patient adherence, and side effects [108]. Future studies should utilize standardized measures over extended periods to determine optimal treatment duration, frequency, and techniques for maintaining benefits [109].

Advances in genomics and neuroimaging could provide biomarkers to guide individualized massage therapy based on a patient’s genetic profile, endogenous opioid function, and baseline neural activity in pain processing regions. This precision approach could identify optimal massage methods and dosing according to the underlying pain mechanisms for each patient [110].

Special considerations should be given to applying massage in vulnerable populations like the elderly, pregnant women, and patients with cancer or recent trauma/fractures [101, 111]. Precautions are needed regarding pressure, positioning, potential drug interactions, and contraindications. Milder massage techniques with supportive aids may be required to ensure safety and tolerability. Overall, an individualized approach factoring patient characteristics is key to maximize massage therapy benefits.

Ultrasound therapy

Ultrasound therapy utilizes high-frequency sound waves to promote tissue healing and relieve chronic pain [112]. It alleviates chronic pain by activating descending inhibitory pathways, modulating neurotransmitters and neuropeptides, reducing inflammation and central sensitization, and stimulating tissue regeneration [113].

In the spinal cord, animal studies suggest ultrasound may inhibit dorsal horn neuron excitability and suppress release of nociceptive neurotransmitters including glutamate, substance P and CGRP, reducing pain signal transmission to the brain [114, 115]. Ultrasound also enhances spinal release of serotonin, norepinephrine, and endogenous opioids that activate descending inhibitory pathways.

Ultrasound may activate cutaneous thermosensitive TRP ion channels to alter peripheral and central pain sensitivity thresholds [116]. Emerging evidence suggests ultrasound increases BDNF, promoting synaptic plasticity and analgesia in pain pathways [117]. Ultrasound therapy may also restore balanced levels of pro- and anti-inflammatory cytokines to mitigate peripheral and central sensitization. It could increase μ-opioid receptor density and decrease inflammatory mediators like cyclooxygenase-2 (COX-2) to reduce nociceptor sensitization [118, 119].

Ultrasound may influence neural plasticity by stimulating neurogenesis, angiogenesis, and synaptogenesis in pain modulatory regions including the periaqueductal gray, hippocampus, and prefrontal cortex [120, 121]. It can promote tissue repair through enhancing cell proliferation, collagen regeneration, and blood flow. Recent studies propose ultrasound may mitigate central sensitization and chronic pain by reducing NMDA receptor activation, neuron excitability, neuroinflammation, and oxidative stress in the spinal cord, thalamus, and pain modulatory areas [122, 123]. Neuroimaging shows ultrasound therapy optimizes functional connectivity in chronic pain-related neural networks [124].

Recent clinical trials demonstrate therapeutic benefits of ultrasound for neuropathic pain conditions like diabetic neuropathy, post-herpetic neuralgia, nerve injury pain, and neck pain [124,125,126,127] (Additional file 1: Fig. S1). The effects may involve modulating nerve excitability, neuroinflammation, and neural plasticity.

For fibromyalgia, ultrasound therapy applied to tender points alleviates widespread pain and sensitivity compared to sham treatment, potentially by stimulating trigger point release and enhancing descending inhibition [128].

As an adjuvant therapy to NSAIDs, membrane stabilizers and opioids, ultrasound demonstrates enhanced analgesia compared to medication alone for chronic back pain, osteoarthritis, and neuropathic pain, possibly through additive sensory-discriminative effects [129,130,131].

Integrating ultrasound into exercise and physiotherapy improves outcomes including pain relief and function versus individual treatments alone for chronic neck, shoulder, and knee pain, optimizing multiple factors influencing chronic pain [128, 132, 133].

While ultrasound therapy demonstrates short-term pain relief, studies evaluating its long-term efficacy and optimal treatment regimens are lacking [134]. There is limited evidence on sustained analgesic effects, ideal dosing frequency and duration, number of sessions required, and prevention of symptom recurrence. Further research through high-quality randomized controlled trials with extended follow-up is warranted to establish protocols maximizing long-term outcomes.

Individual patient factors like age, sex, comorbidities, genetics, baseline pain sensitivity, and endogenous opioid function may influence ultrasound treatment response [113]. Identification of biomarkers predicting treatment outcomes could enable personalized protocols tailored to underlying pain mechanisms. Advances in genomics, neuroimaging, and machine learning techniques could facilitate individualized ultrasound therapy [135].

Potential safety concerns with ultrasound include tissue overheating and cavitation effects. Guidelines on intensity, frequency, duration of application, and monitoring techniques should be followed to avoid tissue damage. Using pulsed ultrasound and keeping transducer movement constant during treatment could improve safety [119]. Overall, individualization of ultrasound therapy while ensuring appropriate treatment parameters is key to optimizing its clinical application.

Light therapy

Light therapy utilizes specific wavelengths of light, such as far infrared and laser, to promote tissue healing and relieve chronic pain [134, 136]. Light therapy demonstrates analgesic effects when applied transcranially to modulate cortical activity as well as when applied locally to the skin surface above painful areas. Light therapy may achieve analgesic effects through mechanisms involving retinal photoreceptors, inflammation, mitochondrial function, and neural activity.

Infrared light may increase nitric oxide and stimulate mitochondrial function, while laser therapy may activate neural pathways mediating analgesia [137, 138]. Light activates retinal opsins and photosensitive proteins that influence neurotransmitters and activity in pain processing brain regions. Light therapy increases BDNF, promoting synaptic plasticity and analgesia. It regulates oxidative stress and inflammation through effects on reactive oxygen species and cytokines [139].

Recent studies propose light therapy may mitigate central sensitization and chronic pain by reducing NMDA receptor activation and neuron excitability in pain pathways [140]. Light also improves functional connectivity between chronic pain-related brain regions including thalamus, somatosensory cortex, insula, cingulate cortex, prefrontal cortex, and periaqueductal gray [139].

Light therapy demonstrates analgesic effects when applied transcranially to modulate cortical activity as well as when applied locally to the skin surface above painful areas [141, 142]. In clinical studies, light therapy demonstrates analgesic effects in diverse chronic pain conditions beyond osteoarthritis, back and neck pain [143, 144]. Recent clinical trials show pain relief with light therapy in fibromyalgia, diabetic neuropathy, chemotherapy-induced peripheral neuropathy, phantom limb pain, post-herpetic neuralgia, and complex regional pain syndrome [145,146,147,148,149,150] (Additional file 1: Fig. S1). The effects may involve modulating peripheral and central neural sensitivity.

As an adjunctive treatment, light therapy combined with pharmacological agents like NSAIDs and opioids shows enhanced analgesia compared to medication alone in randomized trials for low back pain, arthritis pain, and neuropathic pain [151, 152]. Light therapy could exert additive effects through sensory-discriminative pathways.

Integrating light therapy into exercise, physical therapy, and manual therapy programs results in improved outcomes including pain relief and functional gains versus individual treatments alone for chronic musculoskeletal pain disorders. The combination optimizes multiple physiological and neuromodulatory factors influencing chronic pain [134, 153].

In pediatric populations, light therapy reduces pain and discomfort associated with needle-related procedures such as venipuncture and intravenous cannulation based on clinical studies [154]. The mechanisms may involve local vascular and neural effects.

Proper safety precautions should be taken during delivery of light therapy to avoid adverse effects like burns and phototoxicity. Guidelines on intensity, frequency, duration of application, and monitoring techniques should be followed to prevent tissue damage, especially in vulnerable populations [155]. Using intermittent light delivery and monitoring skin temperature could improve safety. Overall, individualization of light therapy while ensuring appropriate treatment parameters is key to optimizing its clinical utility.

Psychological interventions

Psychological interventions are interventions that use cognitive, emotional, or behavioral strategies to modulate pain signals. Psychological interventions can effectively modulate chronic pain through influencing supraspinal pain processing. By altering activity and connectivity in brain regions involved in perception, emotion, cognition, and behavior, such as the thalamus, somatosensory cortex, insula, anterior cingulate cortex, amygdala, prefrontal cortex, and hippocampus, psychological techniques may relieve suffering and restore function.

CBT

CBT is a psychotherapy that aims to change maladaptive thoughts and behaviors related to chronic pain. It works by changing the way people perceive their pain, by challenging negative thoughts and enhancing positive ones through cognitive restructuring and cognitive reappraisal [13]. CBT also targets emotional and behavioral responses to chronic pain, such as fear, anxiety, depression, and anger, by using techniques like exposure therapy, relaxation training, behavioral activation, and emotion regulation [13, 156]. Additionally, CBT addresses lifestyle factors that can impact pain, such as sleep hygiene, diet, physical activity, and social support [156].

Research shows CBT increases endogenous opioid β-endorphin expression and reduces inflammatory factors like IL-6 and TNF-α, suggesting potential analgesic and anti-inflammatory mechanisms [157].

CBT may modulate activity and connectivity of brain regions involved in pain processing, including the prefrontal cortex, anterior cingulate cortex, insula, and amygdala [158]. It regulates pain through “gating” mechanisms, engaging descending modulatory pathways releasing neurotransmitters like opioids, serotonin, and norepinephrine to inhibit spinal nociceptive signaling.

Additionally, CBT improves coping, relaxation, and emotion regulation through techniques like exposure therapy and behavioral activation, targeting pain-related emotions like fear, anxiety, depression, and anger [156].

In addition to low back pain, fibromyalgia, migraine, and neuropathic pain, CBT demonstrates clinical utility across an extensive range of chronic pain disorders [159,160,161]. Recent studies reveal CBT reduces pain and improves function in chronic neck pain, rheumatoid arthritis, irritable bowel syndrome, temporomandibular disorders, chronic abdominal pain, and chronic pelvic pain [9, 162,163,164,165,166] (Additional file 1: Fig. S2). The effects involve altering pain perception, catastrophizing, and coping strategies. For cancer-related pain, CBT combined with pharmacological therapy more effectively reduces pain compared to drug therapy alone, possibly by enhancing pain coping and medication adherence [167].

As an adjuvant treatment, CBT integrated with exercise therapy, massage therapy, and chiropractic care shows greater improvements in pain and function than individual therapies alone for chronic low back pain and osteoarthritis [168]. CBT may have synergistic effects by optimizing sensory, cognitive, and emotional dimensions.

While CBT demonstrates efficacy for chronic pain, long-term benefits and optimal treatment protocols need further research through high-quality randomized controlled trials and longitudinal studies [169]. There is limited evidence on the durability of CBT-induced pain relief and functional gains after treatment cessation. Studies should evaluate sustained effects, ideal session frequency/duration, and value of booster sessions.

Emerging techniques like neuroimaging, EEG, and machine learning may identify biomarkers to guide individualized CBT based on patient characteristics predicting treatment response. For example, functional MRI could indicate baseline neural signatures forecasting CBT outcomes. VR could also be integrated to enhance personalization of CBT content and delivery [170, 171].

CBT is considered very safe with minimal risks. However, comorbid psychological or cognitive deficits may require pre-screening and adapted protocols under supervision to prevent adverse events like anxiety, confusion, or withdrawal [172]. Supportive aids and graded exposure may improve safety and tolerability. Overall CBT demonstrates an excellent safety profile but techniques should be tailored for vulnerable populations.

Hypnosis

Hypnosis is a psychotherapeutic technique that utilizes suggestions to induce a state of altered consciousness, characterized by focused attention, relaxation, and heightened responsiveness to suggestions. This technique operates by altering the perception of pain through modifying the sensory, affective, and cognitive components of chronic pain [173]. Hypnosis also works by modifying the emotional and behavioral responses to chronic pain, by reducing fear, anxiety, depression, and anger, and increasing relaxation, coping, and self-efficacy [174].

Recent neuroimaging studies using functional MRI, positron emission tomography (PET), and EEG reveal hypnosis modulates activity and functional connectivity in multiple brain regions involved in pain perception and modulation, including the anterior cingulate cortex, prefrontal cortex, insula, thalamus, amygdala, and periaqueductal gray [175, 176]. Hypnosis may achieve analgesia by enhancing top-down regulation from cortical and subcortical regions to attenuate ascending nociceptive signaling.

On the molecular level, hypnosis has been found to alter levels of neurotransmitters and neuropeptides involved in pain modulation, including endogenous opioids, serotonin, dopamine, and cholecystokinin [177]. Hypnosis may engage descending inhibitory mechanisms mediated by opioids and monoamines to suppress spinal nociceptive transmission [178].

Recent metabolomics studies indicate hypnosis can regulate various metabolic pathways involved in inflammation, oxidative stress, and energy metabolism that may underlie its analgesic effects. Hypnosis normalizes levels of metabolites related to prostaglandin/leukotriene synthesis, nitric oxide production, and nucleotide metabolism [179].

Emerging epigenetics research indicates hypnosis may exert long-term effects on pain perception by altering chromatin structure and gene expression patterns. Hypnosis induces chromatin remodeling at the promoters of genes encoding pain modulatory factors like opioid receptors, BDNF, and catechol-o-methyltransferase (COMT). This leads to lasting changes in the transcription of these analgesic mediators [180].

Hypnosis demonstrates clinical utility across diverse chronic pain conditions including low back pain, arthritis, fibromyalgia, migraine, cancer pain, and more [181,182,183,184,185] (Additional file 1: Fig. S2). The analgesic effects involve modulating sensory, cognitive, and emotional dimensions of pain.

Importantly, recent evidence highlights the value of hypnosis as an adjunct to optimize pharmacological, physical, and psychological therapies through mechanisms like enhancing descending inhibition, altering pain perception, optimizing attentional processes, and reinforcing positive expectancies and placebo effects [181].

Hypnosis combined with NSAIDs, opioids, and anesthetics demonstrates enhanced analgesia and reduced medication side effects in both acute and chronic pain settings. It improves outcomes of invasive procedures like surgery and childbirth [186]. When integrated with physiotherapy, massage, acupuncture, chiropractic care, and exercise therapy, hypnosis leads to greater improvements in pain, function, and quality of life than individual therapies alone for various musculoskeletal and neuropathic disorders [187].

Hypnosis as an adjuvant to chemotherapy alleviates cancer-related pain and discomfort more effectively than chemotherapy alone [167]. Hypnosis combined with VR therapy demonstrates enhanced analgesia for pediatric acute and procedural pain management [188].

Realizing the full potential of hypnosis will require advances in personalized, precision protocols. Techniques like neuroimaging, EEG, genomics, and machine learning may identify biomarkers to guide individualized hypnosis therapy based on a patient’s genetics, psychology, brain patterns, and outcomes [189]. This could help optimize tailored hypnosis delivery and suggestions for each patient’s needs and pain mechanisms.

Further research should also explore optimal combination strategies for hypnosis as part of multidisciplinary pain management. In particular, studies investigating synergistic effects of hypnosis with newer modalities like VR, brain stimulation, and biologics are warranted. Large collaborative trials evaluating multi-component protocols that integrate hypnosis with pharmacological, psychological and physical therapies will help advance integrative care models.

Mindfulness-based interventions

Mindfulness-based interventions involve using mindful awareness of the present moment without judgment to address pain [190, 191]. These techniques influence the cognitive, affective, and sensory aspects of pain processing.

Neuroimaging studies reveal mindfulness modulates activity and connectivity of key pain-related brain regions like the prefrontal cortex, anterior cingulate cortex, insula, thalamus, and amygdala [192]. Mindfulness may engage top-down regulation from prefrontal cortex and anterior cingulate cortex to inhibit maladaptive pain signaling.

Accumulating evidence indicates mindfulness impacts pain modulatory neurotransmitters including endogenous opioids, endocannabinoids, serotonin, and dopamine [193]. Genomics and proteomics studies indicate mindfulness reduces expression of inflammatory genes and proteins, mitigating peripheral and central sensitization [194].

Mindfulness regulates the autonomic nervous system, restoring sympathetic-parasympathetic balance and normalizing cardiovascular, hypothalamic–pituitary–adrenal axis, and immune system functions involved in chronic stress and pain chronification [195].

Importantly, mindfulness normalizes aberrant connectivity within and between default mode, salience, central executive, and sensorimotor brain networks that underlie cognitive, affective, and sensory processing of pain [196]. This systems-level mechanism provides a neural circuitry framework for mindfulness-based analgesia.

Mindfulness-based interventions demonstrate clinical efficacy across extensive chronic pain disorders beyond fibromyalgia and back pain, including arthritis, headaches, diabetic neuropathy, cancer pain, postoperative pain, irritable bowel syndrome, chronic pelvic pain, temporomandibular joint disorder, and more [197,198,199,200,201,202,203,204,205] (Additional file 1: Fig. S2). The analgesic effects involve modulating cognitive, sensory, and emotional aspects of the multidimensional pain experience.

Notably, mindfulness plus morphine demonstrates greater analgesic effects and fewer side effects than morphine alone for cancer pain, as mindfulness enhances pain coping and expectancies [201]. When incorporated into physiotherapy and massage therapy, mindfulness leads to significantly greater improvements in chronic low back pain than individual therapies alone by optimizing attentional processes [9]. This approach combined with acceptance commitment therapy is more efficacious for reducing arthritis pain and improving mobility than either alone, as the combination reinforces adaptive coping. Additionally, mindfulness plus CBT provides superior migraine symptom relief and quality of life versus either therapy alone by targeting sensory, cognitive and emotional aspects [206].

While promising, there are limitations of mindfulness-based interventions that warrant consideration. Optimal mindfulness protocols tailored to different chronic pain populations and delivery methods need further investigation through high-quality randomized controlled trials and mechanistic studies. Long-term durability of treatment effects, ideal “dosing,” and factors influencing individual variability in response remain unclear. Issues regarding accessibility and patient adherence must also be addressed [207].

Moving forward, several areas require attention to optimize mindfulness-based pain therapies. Development of adapted mindfulness protocols personalized for pediatric, geriatric, and cognitively impaired populations should occur through human-centered design approaches. Optimization of mindfulness training delivery methods, such as VR and smartphone apps, is needed to enhance engagement and accessibility [208, 209]. Further research on mindfulness combination strategies with emerging modalities like neurostimulation, psychedelics, and immunotherapy is essential to develop integrative pain care models.

Overall, continued investigation into mechanisms, personalized protocols, delivery methods, and combination strategies will facilitate unlocking the full potential of mindfulness-based interventions as part of integrative care paradigms for diverse chronic pain disorders.

Placebo effect

Placebo and nocebo effects represent neurophysiological responses that alter pain perception based on expectations and beliefs regarding treatment [210]. Optimizing placebo analgesia and mitigating nocebo hyperalgesia can enhance pain management outcomes. Elucidating the mechanisms of these effects informs strategies to amplify the benefits of pain treatments [210].

Neuroimaging studies using functional MRI reveal placebo and nocebo effects modulate activity and connectivity of key pain modulatory regions like periaqueductal gray, rostral anterior cingulate cortex, prefrontal cortex, amygdala, and hippocampus [211].

PET studies indicate placebo impacts release of endogenous opioid peptides involved in descending inhibition of nociception [212]. Genomics research shows placebo response relates to polymorphisms in genes of the endogenous opioid system [213].

Proteomics analyses demonstrate placebo treatment regulates expression of proteins related to inflammation, neuroplasticity, and oxidative stress. These molecular effects likely contribute to placebo analgesia [214].

Importantly, placebo and nocebo alter aberrant functional connectivity within key brain networks processing sensory, cognitive, and emotional dimensions of pain, including default mode, salience, and central executive networks [15, 211].

Beyond pain models and experimental settings, clinical studies demonstrate placebo treatments provide statistically and clinically meaningful pain relief in extensive chronic pain disorders including low back pain, osteoarthritis, neuropathic pain, fibromyalgia, irritable bowel syndrome, visceral pain, migraine, and tension headaches, among others [211, 215,216,217,218,219,220,221] (Additional file 1: Fig. S2).

Research shows placebo effects have clinical utility when combined with active pharmacological and non-pharmacological therapies for chronic pain. Placebo augmentation of opioid therapy results in greater analgesia at lower drug doses, enabling dose reduction [219]. Placebo combined with NSAIDs or anticonvulsants also enhances pain relief compared to the drugs alone [216]. Additionally, placebo integrated into physiotherapy, massage, acupuncture, mindfulness, and exercise therapy leads to significantly greater improvements in chronic pain versus individual therapies [222].

While promising, there are challenges and limitations in leveraging placebo effects that warrant consideration. The nature of placebo response variability between individuals and unpredictability over time require further elucidation. Ethical concerns regarding deception in placebo administration need addressing [215, 223].

Moving forward, optimizing clinical placebo applications to enhance pain management requires: Standardization of placebo procedures/controls in clinical trials to enable meaningful comparisons. Studies on ethical methods to harness placebo that minimize deception by enhancing positive patient-provider relationships, psychological factors and contextual cues [215]. Investigation of placebo in multimodal approaches combining pharmacotherapy, physical therapy, psychotherapy, and complementary medicine.

Overall, an integrative research framework elucidating the neurobiological, psychological, and ethical dimensions of placebo effects will enable their effective integration into individualized care models to optimize outcomes across diverse pain conditions.

Complementary and alternative therapies

Complementary and alternative therapies are interventions that are not considered part of the conventional medical system, but are used to complement or replace it. Complementary and alternative therapies encompass various techniques that may relieve chronic pain through modulating physiological and psychological factors. Some of the promising options include acupuncture, yoga, tai chi, exercise, and music therapy.

Acupuncture

Acupuncture refers to stimulation of traditional Chinese acupoints. While acupuncture originally involved inserting needles into the body, this section focuses specifically on non-invasive techniques, such as laser or electro-stimulation, to stimulate acupoints. Acupuncture involves stimulating traditional acupoints along meridians to modulate pain signaling. Both manual and electrical stimulation may activate Aδ and C afferent fibers, triggering release of endorphins, enkephalins, and dynorphins that bind to opioid receptors in the periphery, spinal cord, and brain to inhibit nociception [224].

Acupuncture increases local release of adenosine, an endogenous anti-nociceptive agent, which inhibits pain transmission via adenosine A1 receptors based on biochemical assays [225]. Functional MRI reveals acupuncture modulates limbic areas involved in affective pain dimensions and activates anti-nociceptive regions like the periaqueductal gray [226].

Acupuncture elevates central levels of serotonin, norepinephrine, and dopamine, engaging descending inhibitory pathways according to microdialysis studies. Metabolomics analysis indicates acupuncture regulates glucose and lipid metabolism, purine metabolism, and arachidonic acid metabolism involved in anti-inflammatory effects [227, 228].

Emerging evidence suggests acupuncture may reverse central sensitization by reducing NMDA receptor activation, neuroinflammation, and oxidative stress in the spinal cord and pain modulatory brain regions based on immunohistochemistry and polymerase chain reaction (PCR) studies [229].

Clinical studies demonstrate the analgesic effects of acupuncture across diverse chronic pain conditions. Beyond low back pain and knee osteoarthritis, clinical trials show acupuncture analgesic effects in cancer-related pain, dysmenorrhea, migraine, tension headaches, abdominal pain, and chemotherapy-induced peripheral neuropathy, potentially by modulating sensory, affective, and cognitive pain dimensions [230,231,232,233,234,235,236,237] (Additional file 1: Fig. S3).

Importantly, research shows acupuncture enhances chronic pain relief when combined with pharmacological and non-pharmacological therapies. For example, acupuncture plus morphine demonstrates greater analgesic effects and fewer side effects than morphine alone for cancer pain [238]. Acupuncture integrated into physiotherapy programs yields superior improvement in mobility and function compared to either therapy alone for knee osteoarthritis [231]. This highlights the synergistic benefits of acupuncture through modulating multiple mechanisms when combined with other treatment modalities.

While acupuncture demonstrates potential for certain chronic pain conditions, there are limitations warranting consideration [239]. The mechanisms underlying acupuncture analgesia remain incompletely understood. Clinical efficacy varies for different pain disorders and individuals. Sham-controlled studies reveal a large placebo component in acupuncture effects. Safety issues like infection and pneumothorax should be avoided with proper technique and single-use needles.

Moving forward, optimization of acupuncture through computational modeling, neuroimaging, and machine learning could help identify biomarkers predicting response. Integration of electrical and non-invasive laser stimulation may enhance effects while avoiding adverse events. Evidence-based guidelines are needed to inform optimal acupuncture protocols tailored to each chronic pain condition and patient. High-quality randomized controlled trials with extended follow-up should evaluate long-term analgesic effects and impact on function, side effects, and quality of life. Development of acupuncture training programs should emphasize both traditional techniques and emerging evidence [239]. Overall, advancing acupuncture therapy through rigorous research will help unlock its potential within integrative care paradigms for chronic pain management.

Yoga and tai chi

Yoga and tai chi are mind–body practices that may relieve chronic pain and improve wellness through harmonizing physiological and psychological factors. Yoga and tai chi are ancient practices originating from India and China, respectively, which involve physical postures, movements, breathing techniques, and mental focus. They share with modern physical exercise the ability to balance key functions and cultivate optimal pain management [240, 241].

At the molecular level, studies indicate yoga and tai chi increase anti-inflammatory cytokines like IL-10 and IL-4, while reducing pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1β [242, 243]. These effects help counteract chronic inflammatory processes underlying chronic pain conditions.

Functional MRI studies reveal yoga and tai chi can optimize abnormal functional connectivity between key pain-related brain regions. For instance, yoga normalizes altered connectivity between the default mode network and executive control network, which is associated with clinical pain severity [244]. Tai chi improves connectivity between the basal ganglia and sensorimotor cortex, which correlates with restored motor function [245]. Modulating dysfunctional connections in pain-related neural circuitry is crucial for translating neuroimaging findings into clinical utility.

PET imaging demonstrates yoga and tai chi increase release of endogenous opioids, serotonin, and dopamine in pain modulatory pathways including the periaqueductal gray, amygdala, and nucleus accumbens [246]. This activates descending inhibition of nociception.

Advanced EEG signal analysis indicates yoga and tai chi alter theta, alpha, and gamma oscillations associated with sensory, cognitive, and affective processing of pain [244]. This improves neural processing and integration of the pain experience.

Metabolomics studies show yoga and exercise induce changes in metabolites related to energy metabolism, oxidative stress, and inflammation. These biochemical effects may underlie their impact on pain [247, 248].

Epigenetic analyses reveal yoga and tai chi facilitate chromatin remodeling and alter miRNA expression patterns related to pain and analgesia [249]. These lasting changes may mediate long-term impact.

In addition to low back pain and osteoarthritis, emerging clinical evidence demonstrates yoga, tai chi, and physical exercise reduce pain and improve function in other chronic pain conditions, including fibromyalgia, migraine, neuropathic pain, cancer pain, multiple sclerosis pain, and inflammatory bowel disease pain [250,251,252,253,254,255] (Additional file 1: Fig. S3).

For example, several randomized controlled trials show yoga provides clinically meaningful improvements in pain, fatigue, and quality of life in fibromyalgia patients [256]. As an adjunctive therapy, yoga significantly reduces migraine headache frequency, intensity, and disability compared to pharmacotherapy alone [257].

Exercise training alleviates cancer-related pain and chemotherapy-induced peripheral neuropathy based on multiple studies [167]. Tai chi demonstrates significant analgesic effects in peripheral neuropathy patients, enabling reduced use of analgesics [258].

For instance, the combination of yoga and pregabalin is more effective than either therapy alone in reducing neuropathic pain and improving sleep based on a recent randomized controlled trial. Yoga integrated into physiotherapy programs results in superior outcomes compared to individual therapies for chronic low back pain [250]. Mindfulness meditation plus tai chi shows greater improvements in knee osteoarthritis pain and physical function versus either intervention alone [259].

While promising, there are limitations warranting consideration, including unclear optimal protocols for different chronic pain populations, factors influencing individual variability in treatment response, and need for further research on long-term analgesic durability through high-quality longitudinal studies. Accessibility and adherence barriers should also be addressed.

Moving forward, several areas need attention to optimize clinical applications of yoga, tai chi, and exercise for chronic pain. Development of adapted protocols personalized to patient characteristics and needs through predictive algorithms and machine learning is key. Optimization of therapy delivery methods, such as VR, telerehabilitation, and mobile apps, is essential to enhance engagement [260]. Combination strategies with emerging modalities like neurostimulation, nutraceuticals, and biologics should be explored. Large pragmatic trials on multi-component lifestyle programs integrating yoga, tai chi, and exercise are needed to inform integrative care models.

In summary, continued research on mechanisms, predictive biomarkers, delivery methods, and combination approaches will help realize the full potential of yoga, tai chi, and physical exercise as accessible, low-risk interventions that can be integrated into holistic paradigms for chronic pain management.

Music therapy

Music therapy, which employs music interventions to achieve therapeutic goals, encompasses a variety of techniques such as improvisational music therapy, song writing, music performance, and music listening. These techniques may be a promising and safe non-pharmacologic pain management option for patients with chronic pain [261, 262].

Neuroimaging shows music therapy modulates activity and connectivity in areas like thalamus, somatosensory cortex, cingulate cortex, insula, amygdala, prefrontal cortex, and nucleus accumbens [263]. Recent research proposes music therapy may normalize dysfunctional connectivity between default mode, salience, central executive, and reward networks that underlie sensory, affective, and cognitive dimensions of chronic pain [264]. It may reverse maladaptive neuroplasticity and limit central sensitization.

EEG, functional MRI, and computational modeling could help predict individual pain relief response and optimize music therapy protocols. Combining music with pharmacological, physical, or psychological pain treatments may have synergistic effects through peripheral and central mechanisms.

Recent clinical trials demonstrate music therapy decreases pain in additional chronic pain conditions including low back pain, arthritis, migraine headache, chronic neck, and shoulder pain [265,266,267,268] (Additional file 1: Fig. S3). Music alleviates both nociceptive and neuropathic pain through effects on sensory, cognitive, and emotional dimensions.

Integrating music therapy into physical therapy programs improves outcomes including pain relief, mobility, and quality of life versus physical therapy alone for osteoarthritis, rheumatoid arthritis, and chronic neck pain [269]. Music optimizes physiological and psychological influences on chronic pain.

Music therapy combined with CBT enhances pain coping and reduces disability compared to either therapy alone for patients with chronic widespread pain [270]. Music therapy optimizes multiple top-down mechanisms when combined with psychological therapies.

While music therapy demonstrates acute analgesic effects, studies evaluating its long-term efficacy in sustaining pain relief are lacking. Future research should investigate optimal music therapy protocols regarding session frequency, duration, and techniques required for maintaining benefits. Standardized measures over extended periods are needed to determine ideal regimens maximizing enduring outcomes.

Advances in neuroimaging, genetics, and machine learning techniques present opportunities to guide individualized music therapy based on a patient’s brain activity patterns, neurotransmitter function, and music preferences. Computational models could help determine optimal music parameters and predict treatment response. Personalized music selection and delivery may enhance clinical effects [267].

Caution should be taken regarding music characteristics like tempo, rhythm, melody, and volume [271]. Music with abruptly shifting tones or overly stimulating rhythms may exacerbate symptoms in certain chronic pain conditions. Sound levels should be carefully controlled to avoid adverse auditory effects. Overall, individualization of music therapy while considering safety factors can optimize its efficacy as a non-pharmacologic pain management approach.

Others therapies

Additionally, some interventions not classified above also have widespread application in chronic pain management. These methods achieve analgesic effects by acting on different levels of the sensory modulation system.

Manual therapy and movement therapy

Manual therapy refers to hands-on techniques like joint mobilization and soft tissue manipulation aimed at restoring mobility and function [272]. Movement therapy utilizes specific exercises and physical training protocols to improve symptoms [273]. Both approaches influence neural mechanisms across peripheral, spinal, and central levels to alleviate chronic pain.

Manual therapy activates cutaneous mechanoreceptors and modulates spinal processing of sensory signals from the periphery [274]. Movement therapy increases BDNF and synaptic plasticity in pain modulatory circuits [275]. Both normalize imbalances in pro- and anti-inflammatory cytokines linked to chronic pain.

Recent evidence proposes manual therapy and movement therapy may prevent or reverse central sensitization through mechanisms like enhancing descending inhibition, correcting somatic dysfunctions, and optimizing sensory input patterns [276]. Neuroimaging shows normalized functional connectivity in chronic pain-related brain networks after therapy.

EEG and functional MRI have been utilized to evaluate responses and predict outcomes from manual and movement therapies [277]. Combining these approaches with pharmacological, psychological, and other interventions demonstrates enhanced pain relief through synergistic effects on physiological and psychological aspects of chronic pain.

Recent clinical trials demonstrate benefits of manual therapy and exercise for neuropathic pain, fibromyalgia, headache, and temporomandibular joint pain [272, 278,279,280] (Additional file 1: Fig. S4). The effects may involve modulating neural hypersensitivity, inflammation, and muscle dysfunction.

As adjuvants to NSAIDs, membrane stabilizers, and opioids, manual and movement therapies demonstrate enhanced analgesia compared to medication alone for low back pain, neck pain, and osteoarthritis, optimizing both physiological and cognitive-behavioral factors influencing chronic pain [281].

Integrating manual and movement therapies into psychological programs like CBT improves pain coping, catastrophizing, and kinesiophobia compared to either therapy alone [134]. Combination targets both physical dysfunction and maladaptive psychological factors in chronic pain.

While manual therapy and exercise demonstrate short-term pain relief, studies evaluating their long-term effectiveness and optimal protocols are lacking [282]. Future research should investigate the ideal number of sessions, treatment frequency, duration, and techniques required to obtain sustained benefits. Standardized measures over extended periods are needed to determine regimens that maximize enduring outcomes.

Safety considerations for manual therapy include screening for risk factors like osteoporosis and avoiding excessive forces [281]. For movement therapy, progression should be gradual and supervised to prevent overexertion. Special populations like elderly and postoperative patients require modified protocols to ensure safety and tolerability. Overall, individualization along with proper precautions can enhance the safe and effective application of these therapies.

Chiropractic care

Chiropractic care is a healthcare profession that uses manual therapy to treat structural and functional disorders of the spine and joints. Its therapeutic effect on pain relief involves influencing various neural mechanisms involved in pain modulation across different levels of the nervous system [283].

Chiropractic care employs manual techniques to treat musculoskeletal conditions and influence neural mechanisms involved in pain modulation. Chiropractic manipulation activates cutaneous mechanoreceptors and alters spinal processing of peripheral sensory inputs [284]. Emerging evidence suggests chiropractic increases BDNF, facilitating synaptic plasticity in pain pathways [285].

Chiropractic care may mitigate inflammation by reducing pro-inflammatory cytokines and improve circulation through somato-autonomic reflexes [286]. It is proposed to reverse central sensitization by correcting biomechanical and somatic dysfunctions influencing sensory input and motor output patterns.

Neuroimaging studies reveal chiropractic care normalizes aberrant activity and connectivity between chronic pain-related brain regions including the prefrontal cortex, anterior cingulate cortex, insula, and somatosensory cortex [287]. Electrophysiology demonstrates normalized neural hypersensitivity after treatment.

Recent clinical trials demonstrate benefits of chiropractic care for chronic pain conditions like fibromyalgia, headache, neuropathic pain, and temporomandibular disorders [288,289,290,291] (Additional file 1: Fig. S4). The effects may involve modulating neural hypersensitivity, somatic dysfunction, and muscle spasm.

Chiropractic combined with NSAIDs, muscle relaxants, and neuropathic agents demonstrates enhanced analgesia compared to medication alone for low back pain, neck pain, and extremity pain, optimizing the physical and neurological components [292].

Integrating chiropractic care into exercise and physical therapy programs results in improved outcomes including pain relief, range of motion, and function compared to individual therapies alone [293]. The combination targets multiple physiological and biomechanical factors underlying chronic musculoskeletal pain.

Emerging techniques like neuroimaging, genetics, and machine learning may enable individualized chiropractic treatment tailored to each patient’s characteristics and needs [294]. Biomarkers identifying joint dysfunction patterns could guide therapy. Real-time feedback systems could optimize force delivery and dosing for personalized care [294].

Safety considerations include screening for risk factors like osteoporosis, cord compression, and coagulopathies [295]. Excessive manipulation forces and frequencies should be avoided to prevent injury. Special populations like the elderly may require more gentle techniques. Guidelines on application and monitoring should be followed to prevent adverse events. Overall, an individualized approach with proper precaution enhances chiropractic treatment safety and efficacy.

Biofeedback

Biofeedback is a non-invasive technique that uses sensors to measure physiological signals such as heart rate, muscle tension, skin temperature, or brain activity and provides real-time feedback to help individuals develop awareness and control over these signals [296]. When combined with psychosocial or physical therapies, biofeedback may help relieve chronic pain by altering bodily functions and influencing pain-related brain networks [297, 298].

It should be noted that biofeedback is rarely used as a standalone intervention, but rather integrated with other physical and psychological therapies. The mechanisms of biofeedback should be understood in light of its synergistic effects when combined with modalities like exercise, massage, CBT, and more. Focusing solely on the mechanisms of biofeedback itself risks decontextualizing it from real-world, multimodal applications.

Integrating biofeedback into exercise, massage, and physical therapy programs results in greater improvements in pain, mobility, and quality of life versus either therapy alone for musculoskeletal pain, temporomandibular joint disorders, irritable bowel syndrome, chronic fatigue syndrome, and pelvic pain [299,300,301,302,303,304] (Additional file 1: Fig. S4).

Combining biofeedback with CBT, mindfulness, and hypnosis therapy shows greater reductions in pain-related anxiety, fear avoidance, and catastrophizing thoughts compared to individual treatments [305]. Multimodal approaches utilizing biofeedback may achieve optimal pain self-regulation.

Caution should be taken with electrode placement and stimulus intensity during biofeedback. Patients with cardiac devices or seizures require modified protocols [306]. Guidance on use and monitoring should be provided, especially for home-based treatment. Combining self-regulation techniques with biofeedback may optimize skills retention and adherence.

Future studies could examine innovative biofeedback modalities, remote delivery methods, and integration with VR or gaming for engagement. Big data analytics and AI could uncover new strategies to enhance pain self-management through biofeedback [307]. Overall, advancing biofeedback technology while ensuring appropriate applications can maximize its potential as a chronic pain treatment.

VR therapy

VR therapy utilizes computer-generated environments to create immersive experiences for chronic pain relief [308]. VR therapy may achieve analgesia through multiple mechanisms, including cognitively distractive effects, visuotactile stimulation, and visuomotor stimulation [309, 310]. These multimodal effects may induce neuroplastic changes by altering sensory perception and modulating pain-related neural processing. Studies suggest that VR therapy can induce analgesia, modulate neural plasticity, and alter brain activity related to anxiety disorders.

Emerging evidence suggests VR therapy may prevent or reverse central sensitization, a key mechanism in the transition from acute to chronic pain [311]. At the molecular level, VR therapy reduces NMDA receptor activation and neuropeptide release involved in spinal cord hyperexcitability based on animal studies. It normalizes elevated COX-2, TNF-α, and IL-1β levels associated with neuroinflammation in pain pathways [312]. At the cellular level, VR therapy decreases dorsal horn neuron firing and wind-up in response to repeated stimuli. It suppresses long-term potentiation of synaptic transmission between nociceptive afferents and spinal projection neurons [313]. These effects indicate VR therapy may inhibit the molecular and cellular processes driving central sensitization. Clinically, VR applied early after injury prevents chronic pain development compared to conventional treatment in human studies. Overall, VR therapy holds promise for inhibiting maladaptive neuroplasticity underlying the progression from acute to chronic pain through counteracting the key mechanisms of central sensitization.

Neuroimaging reveals VR therapy activates endogenous anti-nociception systems like the periaqueductal gray and nucleus raphe magnus, which engage descending inhibition of nociceptive signals [314]. Functional MRI shows VR therapy enhances functional connectivity between periaqueductal gray and pain-processing cortical regions to strengthen top-down pain modulation [315].

VR therapy facilitates neuroplastic changes in brain networks involved in sensory, affective and cognitive processing of chronic pain. Resting-state functional MRI indicates VR normalizes aberrant default mode network connectivity associated with clinical pain [315]. Diffusion MRI shows VR alters structural connectivity between thalamus, periaqueductal gray, and sensory cortices critical for pain transmission [316].

Recent clinical trials demonstrate VR analgesic effects in diverse chronic pain populations beyond neuropathic and postoperative pain. In fibromyalgia, VR reduces pain scores and increases threshold compared to control treatment [317]. For migraine, VR adjunctive to pharmacologic prophylaxis leads to greater reductions in headache frequency and disability than drug therapy alone [318]. In cancer pain, VR combined with opioids provides superior analgesia at lower opioid doses versus opioids alone, highlighting the broad applicability of VR across different chronic pain conditions [319] ( Additional file 1: Fig. S4).

Importantly, research shows VR has synergistic benefits when combined with pharmacological and non-pharmacological treatments. For example, VR plus pregabalin reduces neuropathic pain and pregabalin-associated somnolence more than either treatment alone [320]. VR integrated into physiotherapy yields greater improvements in function compared to individual therapies for chronic low back pain [321]. VR adjunctive to CBT enhances pain coping and reduces anxiety better than either therapy alone [322]. Such combinatorial approaches facilitate multimodal engagement of sensory, cognitive, and emotional aspects to optimize outcomes. Overall, these studies highlight VR as a promising adjuvant that can amplify analgesic effects within multidisciplinary chronic pain care.

While VR therapy shows promise, there are limitations needing consideration. Individual differences in immersion and side effects like cybersickness can influence treatment response [323]. For patients with allodynia or hyperalgesia, visuotactile and visuomotor content requires caution as it may exacerbate symptoms [324]. In these cases, individualized approaches avoiding tactile or motor stimulation may be necessary. Moving forward, advances in VR technology and machine learning could enhance personalization and accessibility. For example, AI and neuroimaging may reveal biomarkers to optimize protocols on an individualized basis. Multisensory simulations tailored to specific pain conditions and customized triggers could improve therapeutic benefits. Through human-centered design and integrating engineering innovations with neuroscience insights, VR therapy can achieve its full potential as an effective personalized treatment for chronic pain.

Non-invasive brain stimulation (NIBS)

NIBS utilizes magnetic or electric fields to modulate brain activity for pain relief. Different NIBS techniques have different mechanisms of action on pain-related neural circuits [325]. It is postulated that NIBS such as transcranial magnetic stimulation (TMS) and transcranial direct current stimulation (tDCS) modulates cortical excitability and activity to induce analgesia.

Spectroscopy studies show NIBS alters levels of neurotransmitters including glutamate, GABA, serotonin, and dopamine critical for pain processing [325]. Proteomics analysis indicates NIBS modulates expression of receptors like NMDA, opioid, and TRPV1 involved in nociception [326].

Neuroimaging studies reveal the molecular effects of NIBS manifest as changes in pain-related neural circuitry [327]. PET imaging indicates NIBS engages descending pain inhibitory pathways including the periaqueductal gray, rostral ventromedial medulla, and opioidergic nucleus cuneiformis [328]. Functional MRI reveals NIBS normalizes aberrant activity and connectivity in the default mode network, salience network, and sensorimotor network which underlie cognitive, affective, and sensory dimensions of chronic pain [329].

NIBS facilitates neuroplasticity through long-term potentiation and depression of synaptic strength. Structural MRI shows NIBS induces gray matter changes in pain-related regions including thalamus, somatosensory cortex, anterior cingulate cortex, and insula [330].

Emerging NIBS techniques like temporal interference stimulation, transcranial pulsed ultrasound, and transcutaneous vagus nerve stimulation target peripheral nerves and the vagus nerve to engage endogenous analgesia mechanisms [331, 332]. Temporal interference stimulation works by delivering out-of-phase currents to the head which affects pain-related cortical areas [333]. Transcranial pulsed ultrasound modulates brain circuits through mechanical and thermal effects [331]. Vagus nerve stimulation activates the vagus nerve’s connection to the locus coeruleus and pain modulatory nuclei [332]. These novel techniques expand the scope of NIBS for non-invasive pain relief.

Recent studies demonstrate analgesic effects of NIBS in diverse chronic pain populations beyond neuropathic pain and headaches, including cancer-related pain, visceral pain, abdominal pain, fibromyalgia, and musculoskeletal pain [334,335,336,337,338] (Additional file 1: Fig. S4). NIBS also reduces acute postoperative pain and opioid consumption. These findings highlight the wide applicability of NIBS for pain management.

Importantly, NIBS demonstrates enhanced pain relief when combined with pharmacological and non-pharmacological therapies. For instance, NIBS applied before motor cortex stimulation improves treatment outcomes for neuropathic pain. NIBS combined with exercise therapy yields greater gains in pain and function compared to either therapy alone for fibromyalgia [339]. This highlights NIBS as a promising adjuvant that can amplify analgesic effects and improve clinical outcomes when combined with other treatment modalities as part of multidisciplinary care.

While promising, there are limitations of NIBS warranting consideration. The mechanisms underlying different techniques remain incompletely understood [340]. Response variability between patients and unpredictable effects over time require further study. Safety and tolerability depend on stimulus parameters and individual factors. Headaches, fatigue, nausea, insomnia, and pain exacerbation can sometimes occur. Special consideration is required for vulnerable populations like pediatric, elderly, and pregnant patients.

Moving forward, advances in computational modeling and machine learning applied to neuroimaging, electrophysiological, genetic, and clinical data could help optimize personalized NIBS protocols tailored to each patient’s symptoms, biomarkers, and responses. Technological improvements such as novel electrode designs, wearable devices, and integration with function-triggered feedback systems could enhance individualized delivery, safety, and outcomes. Development of NIBS protocols adapted for home use with telemonitoring may increase accessibility. Further research on combination strategies with emerging treatments like immunomodulation, VR therapy, and psychedelics could uncover synergies [341]. Overall, continued innovation and optimization of NIBS technology and protocols through a precision medicine approach will help fulfill its potential as a safe, effective personalized therapy for diverse chronic pain disorders.

Summary: common mechanisms of major NINPT modalities

Now that we have reviewed the mechanisms of major NINPT modalities respectively, it is worth summarizing some of their common principles from a comparative perspective. While NINPT modalities differ in their techniques, they share some common mechanisms that confer analgesic effects:

  • Modulating nociceptive signaling—Most NINPT can inhibit pain signal transmission in the peripheral and central nervous system through effects on ion channels, neurotransmitters, and inflammation.

  • Enhancing descending modulation—Many NINPT engage endogenous analgesic pathways involving opioids, serotonin, and norepinephrine to exert inhibitory effects.

  • Influencing psychology and emotions—NINPT like CBT and hypnosis target affective and cognitive dimensions of pain.

  • Promoting neuroplasticity—Some NINPT may facilitate neuroplastic changes in pain pathways through mechanisms like long-term potentiation (LTP)/long-term depression (LTD).

  • Limiting sensitization—Certain NINPT are proposed to prevent or reverse central sensitization underlying chronic pain.

Although the specific molecular and cellular effects vary across techniques, summarizing these common principles may provide a preliminary framework to guide optimal application and combination of different NINPT modalities. Despite current differences, the common mechanisms of major NINPT modalities highlighted here represent our initial attempts to inform the selection and integration of appropriate techniques for individualized, multimodal chronic pain management.

Limitations and challenges in the use of NINPT

While NINPT demonstrates potential for managing pain without medications or invasive procedures, there are limitations warranting consideration.

Heterogeneity and individualization

The heterogeneity of chronic pain conditions and individual variability in treatment response highlight the need to tailor NINPT protocols based on each patient’s specific needs and mechanisms [342,343,344]. Treatment outcomes should be monitored and adjusted accordingly. Advances in predictive biomarkers could guide individualized approaches.

Standardization

The lack of standardized treatment protocols and dosing guidelines makes it difficult to compare efficacy and generalize findings across different NINPT modalities and trials [345, 346]. Validated, reliable outcome measures are needed to quantify neural activity changes and pain relief. Computational modeling and machine learning may help optimize protocols.

Accessibility and adherence

Practical barriers like affordability, insurance coverage, and access to specialized equipment or facilities may limit utilization of certain NINPT. Patient adherence may suffer if expectations are not properly managed. Cost-effectiveness studies and enhanced engagement strategies are required to promote adherence and accessibility [347].

Research methodology limitations

Current studies exhibit issues like small sample sizes, inadequate randomization, blinding difficulties, and lack of long-term follow-up. Research gaps exist in elucidating mechanisms and predictive biomarkers. Pragmatic comparative trials of different NINPT protocols are needed. Addressing these limitations through rigorous research is key to inform clinical translation and optimize patient outcomes [348].

Future outlook and recommendations

To realize the full potential of NINPT, further innovation and optimization is needed across several fronts:

Advancing personalized and precision therapies

Emerging techniques utilizing computational modeling, neuroimaging, omics, and machine learning have proposed personalized medicine approaches for chronic pain treatment [349,350,351]. Further integration of these technologies could facilitate developing individualized NINPT protocols tailored to each patient’s specific symptoms, predictive biomarkers, and treatment responses. Additionally, combining NINPT with nutrition and lifestyle interventions may also help optimize therapeutic outcomes.

Promoting integrative and combinatorial treatments

Synergistic combination strategies that incorporate NINPT modalities with pharmacological, psychological, physical, and complementary interventions are investigated through a systems approach [352, 353]. This multimodal integration can better engage pain mechanisms at peripheral, spinal, and supraspinal levels.

Harnessing emerging technologies and neuroscience insights

Applying biosensors, wearables, and implantables enables adaptive closed-loop NINPT delivery [354, 355]. Utilizing advanced techniques like optogenetics, holographic microscopy, and functional neuroimaging to elucidate NINPT mechanisms across multiple levels of analysis.

Improving research methodology

Addressing limitations in study design, standardization, comparator selection, and long-term assessments through pragmatic, large-scale comparative trials focused on patient-centered outcomes [356, 357]. This will strengthen the evidence base to inform guidelines for clinically translating different NINPT.

Conclusions

NINPT represents a promising domain encompassing diverse interventions that modulate pain through peripheral, spinal, and supraspinal mechanisms. This review synthesizes current evidence on mechanisms and clinical potential of major NINPT modalities. While results are promising, limitations warrant attention (Fig. 4).

Fig. 4
figure 4

Overview of non-pharmacological therapies for chronic pain. Note: TENS: transcutaneous electrical nerve stimulation; CBT: cognitive-behavioral therapy; VR: virtual reality

Realizing the full benefits of NINPT requires a personalized, integrative approach combining technologies, research innovations, and therapeutic strategies across pharmacological, psychological, and physical domains. Advancing NINPT through translational research and addressing methodology limitations will provide the evidence base to inform clinical guidelines and integrate NINPT into precision care models.

Overall, this review bridges the gap between neuroscience insights and clinical translation to highlight the value of NINPT as a complementary non-pharmacological option ready to be tailored to individual needs and integrated into contemporary pain medicine. We hope this mechanistic framework promotes implementation of NINPT in clinical practice to ultimately improve outcomes and quality of life for chronic pain patients.

Availability of data and materials

Not applicable.

Abbreviations

ACC:

Anterior cingulate cortex

BDNF:

Brain-derived neurotrophic factor

CBT:

Cognitive-behavioral therapy

COMT:

Catechol-o-methyltransferase

COX-2:

Cyclooxygenase-2

EEG:

Electroencephalogram

IC:

Insula cortex

IL-6:

Interleukin-6

MRI:

Magnetic resonance imaging

NIBS:

Non-invasive brain stimulation

NINPT:

Non-invasive non-pharmacological therapies

NSAIDs:

Nonsteroidal anti-inflammatory drugs

PAG:

Periaqueductal gray

PCR:

Polymerase chain reaction

PET:

Positron emission tomography

PFC:

Prefrontal cortex

RVM:

Rostral ventromedial medulla

tDCS:

Transcranial direct current stimulation

TENS:

Transcutaneous electrical nerve stimulation

TMS:

Transcranial magnetic stimulation

TNF-α:

Tumor necrosis factor alpha

VR:

Virtual reality

References

  1. Yong RJ, Mullins PM, Bhattacharyya N. Prevalence of chronic pain among adults in the United States. Pain. 2022;163(2):e328–32.

    PubMed  Google Scholar 

  2. Kuehn B. Chronic pain prevalence. JAMA. 2018;320(16):1632–1632.

    PubMed  Google Scholar 

  3. Mills S, Torrance N, Smith BH. Identification and management of chronic pain in primary care: a review. Curr Psychiatry Rep. 2016;18:1–9.

    Google Scholar 

  4. Reid MC, Eccleston C, Pillemer K. Management of chronic pain in older adults. BMJ. 2015;350:h532.

    PubMed  PubMed Central  Google Scholar 

  5. Chou R, Fanciullo GJ, Fine PG, Adler JA, Ballantyne JC, Davies P, Donovan MI, Fishbain DA, Foley KM, Fudin J. Clinical guidelines for the use of chronic opioid therapy in chronic noncancer pain. J Pain. 2009;10(2):113-30.e22.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Fowler TO, Durham CO, Planton J, Edlund BJ. Use of nonsteroidal anti-inflammatory drugs in the older adult. J Am Assoc Nurse Pract. 2014;26(8):414–23.

    PubMed  Google Scholar 

  7. Wolkerstorfer A, Handler N, Buschmann H. New approaches to treating pain. Bioorg Med Chem Lett. 2016;26(4):1103–19.

    CAS  PubMed  Google Scholar 

  8. Qaseem A, Wilt TJ, McLean RM, Forciea MA, Denberg TD, Barry MJ, Boyd C, Chow RD, Fitterman N, Harris RP, et al. Noninvasive treatments for acute, subacute, and chronic low back pain: a clinical practice guideline from the American College of Physicians. Ann Intern Med. 2017;166(7):514–30.

    PubMed  Google Scholar 

  9. Skelly AC, Chou R, Dettori JR, Turner JA, Friedly JL, Rundell SD, Fu R, Brodt ED, Wasson N, Kantner S, et al. Noninvasive nonpharmacological treatment for chronic pain: a systematic review update. Agency for Healthcare Research and Quality (US): Rockville; 2020.

    Google Scholar 

  10. Johnson MI, Paley CA, Jones G, Mulvey MR, Wittkopf PG. Efficacy and safety of transcutaneous electrical nerve stimulation (TENS) for acute and chronic pain in adults: a systematic review and meta-analysis of 381 studies (the meta-TENS study). BMJ Open. 2022;12(2):e051073.

    PubMed  PubMed Central  Google Scholar 

  11. Wang Y, Lu H, Li S, Zhang Y, Yan F, Huang Y, Chen X, Yang A, Han L, Ma Y. Effect of cold and heat therapies on pain relief in patients with delayed onset muscle soreness: a network meta-analysis. J Rehabil Med. 2022;54:jrm00258.

    PubMed  Google Scholar 

  12. Field T. Massage therapy research review. Complement Ther Clin Pract. 2016;24:19–31.

    PubMed  PubMed Central  Google Scholar 

  13. Ehde DM, Dillworth TM, Turner JA. Cognitive-behavioral therapy for individuals with chronic pain: efficacy, innovations, and directions for research. Am Psychol. 2014;69(2):153.

    PubMed  Google Scholar 

  14. Cherkin DC, Sherman KJ, Balderson BH, Cook AJ, Anderson ML, Hawkes RJ, Hansen KE, Turner JA. Effect of mindfulness-based stress reduction vs cognitive behavioral therapy or usual care on back pain and functional limitations in adults with chronic low back pain: a randomized clinical trial. JAMA. 2016;315(12):1240–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Shi Y, Cui S, Zeng Y, Huang S, Cai G, Yang J, Wu W. Brain network to placebo and nocebo responses in acute experimental lower back pain: a multivariate granger causality analysis of fMRI data. Front Behav Neurosci. 2021;15:696577.

    PubMed  PubMed Central  Google Scholar 

  16. Shi Y, Liu Z, Zhang S, Li Q, Guo S, Yang J, Wu W. Brain network response to acupuncture stimuli in experimental acute low back pain: an fMRI study. Evid-Based Compl Alt: eCAM. 2015;2015:210120.

    Google Scholar 

  17. Saper RB, Lemaster C, Delitto A, Sherman KJ, Herman PM, Sadikova E, Stevans J, Keosaian JE, Cerrada CJ, Femia AL, et al. Yoga, physical therapy, or education for chronic low back pain: a randomized noninferiority trial. Ann Intern Med. 2017;167(2):85–94.

    PubMed  PubMed Central  Google Scholar 

  18. Gutgsell KJ, Schluchter M, Margevicius S, DeGolia PA, McLaughlin B, Harris M, Mecklenburg J, Wiencek C. Music therapy reduces pain in palliative care patients: a randomized controlled trial. J Pain Symptom Manage. 2013;45(5):822–31.

    PubMed  Google Scholar 

  19. Urits I, Schwartz RH, Orhurhu V, Maganty NV, Reilly BT, Patel PM, Wie C, Kaye AD, Mancuso KF, Kaye AJ. A comprehensive review of alternative therapies for the management of chronic pain patients: acupuncture, tai chi, osteopathic manipulative medicine, and chiropractic care. Adv Ther. 2021;38:76–89.

    PubMed  Google Scholar 

  20. Bergmann A, Edelhoff D, Schubert O, Erdelt KJ, Pho Duc JM. Effect of treatment with a full-occlusion biofeedback splint on sleep bruxism and TMD pain: a randomized controlled clinical trial. Clin Oral Investig. 2020;24(11):4005–18.

    PubMed  PubMed Central  Google Scholar 

  21. Ahmadpour N, Randall H, Choksi H, Gao A, Vaughan C, Poronnik P. Virtual Reality interventions for acute and chronic pain management. Int J Biochem Cell Biol. 2019;114:105568.

    CAS  PubMed  Google Scholar 

  22. Peng WW, Tang ZY, Zhang FR, Li H, Kong YZ, Iannetti GD, Hu L. Neurobiological mechanisms of TENS-induced analgesia. Neuroimage. 2019;195:396–408.

    CAS  PubMed  Google Scholar 

  23. Medaglia JD, Erickson B, Zimmerman J, Kelkar A. Personalizing neuromodulation. Int J Psychophysiol. 2020;154:101–10.

    PubMed  Google Scholar 

  24. Bannister K, Sachau J, Baron R, Dickenson AH. Neuropathic pain: mechanism-based therapeutics. Annu Rev Pharmacol Toxicol. 2020;60:257–74.

    CAS  PubMed  Google Scholar 

  25. Corp N, Mansell G, Stynes S, Wynne-Jones G, Morsø L, Hill JC, van der Windt DA. Evidence-based treatment recommendations for neck and low back pain across Europe: a systematic review of guidelines. Eur J Pain. 2021;25(2):275–95.

    PubMed  Google Scholar 

  26. Apkarian AV, Bushnell MC, Treede RD, Zubieta JK. Human brain mechanisms of pain perception and regulation in health and disease. Eur J Pain. 2005;9(4):463–84.

    PubMed  Google Scholar 

  27. Fornasari D. Pain mechanisms in patients with chronic pain. Clin Drug Investig. 2012;32:45–52.

    CAS  PubMed  Google Scholar 

  28. Feizerfan A, Sheh G. Transition from acute to chronic pain. Contin Educ Anaesth Crit Care Pain. 2015;15(2):98–102.

    Google Scholar 

  29. Borsook D. Neurological diseases and pain. Brain. 2011;135(2):320–44.

    PubMed  PubMed Central  Google Scholar 

  30. Denk F, Bennett DL, McMahon SB. Nerve growth factor and pain mechanisms. Annu Rev Neurosci. 2017;40:307–25.

    CAS  PubMed  Google Scholar 

  31. Dubner R, Hargreaves KM. The neurobiology of pain and its modulation. Clin J Pain. 1989;5(2):S1-6.

    PubMed  Google Scholar 

  32. Qiao LN, Liu JL, Tan LH, Yang HL, Zhai X, Yang YS. Effect of electroacupuncture on thermal pain threshold and expression of calcitonin-gene related peptide, substance P and γ-aminobutyric acid in the cervical dorsal root ganglion of rats with incisional neck pain. Acupunct Med. 2017;35(4):276–83.

    PubMed  PubMed Central  Google Scholar 

  33. Otsuka M, Yanagisawa M. Pain and neurotransmitters. Cell Mol Neurobiol. 1990;10(3):293–302.

    CAS  PubMed  Google Scholar 

  34. Gamal-Eltrabily M, Martínez-Lorenzana G, González-Hernández A, Condés-Lara M. Cortical modulation of nociception. Neuroscience. 2021;458:256–70.

    CAS  PubMed  Google Scholar 

  35. Tracey I, Mantyh PW. The cerebral signature for pain perception and its modulation. Neuron. 2007;55(3):377–91.

    CAS  PubMed  Google Scholar 

  36. Woolf CJ. Central sensitization: implications for the diagnosis and treatment of pain. Pain. 2011;152(3):S2–15.

    PubMed  Google Scholar 

  37. Hemington KS, Coulombe M-A. The periaqueductal gray and descending pain modulation: why should we study them and what role do they play in chronic pain? J Neurophysiol. 2015;114(4):2080–3.

    PubMed  Google Scholar 

  38. Fuchs PN, Peng YB, Boyette-Davis JA, Uhelski ML. The anterior cingulate cortex and pain processing. Front Integr Neurosci. 2014;8:35.

    PubMed  PubMed Central  Google Scholar 

  39. Ong W-Y, Stohler CS, Herr DR. Role of the prefrontal cortex in pain processing. Mol Neurobiol. 2019;56:1137–66.

    CAS  PubMed  Google Scholar 

  40. Vardeh D, Mannion RJ, Woolf CJ. Toward a mechanism-based approach to pain diagnosis. J Pain. 2016;17(9):T50–69.

    PubMed  PubMed Central  Google Scholar 

  41. Sanzarello I, Merlini L, Rosa MA, Perrone M, Frugiuele J, Borghi R, Faldini C. Central sensitization in chronic low back pain: a narrative review. J Back Musculoskelet Rehabil. 2016;29(4):625–33.

    PubMed  Google Scholar 

  42. Trouvin AP, Perrot S. New concepts of pain. Best Pract Res Clin Rheumatol. 2019;33(3):101415.

    PubMed  Google Scholar 

  43. Coderre TJ, Katz J, Vaccarino AL, Melzack R. Contribution of central neuroplasticity to pathological pain: review of clinical and experimental evidence. Pain. 1993;52(3):259–85.

    CAS  PubMed  Google Scholar 

  44. Woolf CJ, Salter MW. Neuronal plasticity: increasing the gain in pain. Science. 2000;288(5472):1765–8.

    CAS  PubMed  Google Scholar 

  45. Bliss TV, Collingridge GL, Kaang B-K, Zhuo M. Synaptic plasticity in the anterior cingulate cortex in acute and chronic pain. Nat Rev Neurosci. 2016;17(8):485–96.

    CAS  PubMed  Google Scholar 

  46. Vachon-Presseau E, Tétreault P, Petre B, Huang L, Berger SE, Torbey S, Baria AT, Mansour AR, Hashmi JA, Griffith JW, et al. Corticolimbic anatomical characteristics predetermine risk for chronic pain. Brain. 2016;139(7):1958–70.

    PubMed  PubMed Central  Google Scholar 

  47. Nijs J, Leysen L, Vanlauwe J, Logghe T, Ickmans K, Polli A, Malfliet A, Coppieters I, Huysmans E. Treatment of central sensitization in patients with chronic pain: time for change? Expert Opin Pharmaco. 2019;20(16):1961–70.

    Google Scholar 

  48. Liebano RE, Rakel B, Vance CGT, Walsh DM, Sluka KA. An investigation of the development of analgesic tolerance to TENS in humans. Pain. 2011;152(2):335–42.

    PubMed  Google Scholar 

  49. Mokhtari T, Ren Q, Li N, Wang F, Bi Y, Hu L. Transcutaneous electrical nerve stimulation in relieving neuropathic pain: basic mechanisms and clinical applications. Curr Pain Headache Rep. 2020;24(4):14.

    PubMed  Google Scholar 

  50. Wu LC, Weng PW, Chen CH, Huang YY, Tsuang YH, Chiang CJ. Literature review and meta-analysis of transcutaneous electrical nerve stimulation in treating chronic back pain. Region Anesth Pain M. 2018;43(4):425–33.

    Google Scholar 

  51. Shimo K, Fukui H, Ueta H, Naka M, Miyamoto S, Nishikawa E, Ohga S, Matsubara T. Decrease in current perception thresholds of A-beta fibers by subthreshold noise stimulation using transcutaneous electrical nerve stimulation. NeuroReport. 2023;34(5):287–9.

    PubMed  Google Scholar 

  52. Krabbenbos IP, Brandsma D, van Swol CF, Boezeman EH, Tromp SC, Nijhuis HJ, van Dongen EP. Inhibition of cortical laser-evoked potentials by transcutaneous electrical nerve stimulation. Neuromodulation. 2009;12(2):141–5.

    PubMed  Google Scholar 

  53. Lin H-T, Chiu C-C, Wang J-J, Hung C-H, Chen Y-W. High frequency transcutaneous electrical nerve stimulation with diphenidol administration results in an additive antiallodynic effect in rats following chronic constriction injury. Neurosci Lett. 2015;589:62–6.

    CAS  PubMed  Google Scholar 

  54. Sabino GS, Santos CM, Francischi JN, De Resende MA. Release of endogenous opioids following transcutaneous electric nerve stimulation in an experimental model of acute inflammatory pain. J Pain. 2008;9(2):157–63.

    CAS  PubMed  Google Scholar 

  55. Rokugo T, Takeuchi T, Ito H. A histochemical study of substance P in the rat spinal cord: effect of transcutaneous electrical nerve stimulation. J Nippon Med Sch. 2002;69(5):428–33.

    CAS  PubMed  Google Scholar 

  56. Garrison DW, Foreman RD. Decreased activity of spontaneous and noxiously evoked dorsal horn cells during transcutaneous electrical nerve stimulation (TENS). Pain. 1994;58(3):309–15.

    PubMed  Google Scholar 

  57. Chandran P, Sluka KA. Development of opioid tolerance with repeated transcutaneous electrical nerve stimulation administration. Pain. 2003;102(1–2):195–201.

    CAS  PubMed  Google Scholar 

  58. Hingne PM, Sluka KA. Blockade of NMDA receptors prevents analgesic tolerance to repeated transcutaneous electrical nerve stimulation (TENS) in rats. J Pain. 2008;9(3):217–25.

    CAS  PubMed  Google Scholar 

  59. Choi J, Kim J, Kang E, Lee JM, Cha J, Kim Y, Lee H, Choi JH, Yi DJ. Brain mechanisms of pain relief by transcutaneous electrical nerve stimulation: a functional magnetic resonance imaging study. Eur J Pain. 2016;20(1):92–105.

    CAS  PubMed  Google Scholar 

  60. Avantaggiato F, Farokhniaee A, Bandini A, Palmisano C, Hanafi I, Pezzoli G, Mazzoni A, Isaias IU. Intelligibility of speech in Parkinson’s disease relies on anatomically segregated subthalamic beta oscillations. Neurobiol Dis. 2023;1:106239.

    Google Scholar 

  61. Santuzzi CH, Neto HdAF, Pires JGP, Gonçalves WLS, Gouvea SA, Abreu GR. High-frequency transcutaneous electrical nerve stimulation reduces pain and cardio-respiratory parameters in an animal model of acute pain: participation of peripheral serotonin. Physiother Theory Pract. 2013;29(8):630–8.

    PubMed  Google Scholar 

  62. Gürgen SG, Sayın O, Çetin F, Tuç YA. Transcutaneous electrical nerve stimulation (TENS) accelerates cutaneous wound healing and inhibits pro-inflammatory cytokines. Inflammation. 2014;37:775–84.

    PubMed  Google Scholar 

  63. Johnson MI, Bjordal JM. Transcutaneous electrical nerve stimulation for the management of painful conditions: focus on neuropathic pain. Expert Rev Neurother. 2011;11(5):735–53.

    PubMed  Google Scholar 

  64. Jin DM, Xu Y, Geng DF, Yan TB. Effect of transcutaneous electrical nerve stimulation on symptomatic diabetic peripheral neuropathy: a meta-analysis of randomized controlled trials. Diabetes Res Clin Pract. 2010;89(1):10–5.

    PubMed  Google Scholar 

  65. Stepanović A, Kolšek M, Kersnik J, Erčulj V. Prevention of post-herpetic neuralgia using transcutaneous electrical nerve stimulation. Wien Klin Wochenschr. 2015;127(9):369–74.

    PubMed  Google Scholar 

  66. Özkul Ç, Kılınç M, Yıldırım SA, Topçuoğlu EY, Akyüz M. Effects of visual illusion and transcutaneous electrical nerve stimulation on neuropathic pain in patients with spinal cord injury: a randomised controlled cross-over trial. J Back Musculoskelet Rehabil. 2015;28(4):709–19.

    PubMed  Google Scholar 

  67. Tonye-Geoffroy L, Mauboussin Carlos S, Tuffet S, Fromentin H, Berard L, Leblanc J, Laroche F. Efficacy of a combination of hypnosis and transcutaneous electrical nerve stimulation for chronic non-cancer pain: a randomized controlled trial. J Adv Nurs. 2021;77(6):2875–86.

    PubMed  PubMed Central  Google Scholar 

  68. Shi X, Yu W, Zhang W, Wang T, Battulga O, Wang L, Guo C. A comparison of the effects of electroacupuncture versus transcutaneous electrical nerve stimulation for pain control in knee osteoarthritis: a Bayesian network meta-analysis of randomized controlled trials. Acupunct Med. 2021;39(3):163–74.

    PubMed  Google Scholar 

  69. Brosseau L, Yonge K, Marchand S, Robinson V, Osiri M, Wells G, Tugwell P. Efficacy of transcutaneous electrical nerve stimulation for osteoarthritis of the lower extremities: a meta-analysis. Phys Ther Rev. 2004;9(4):213–33.

    Google Scholar 

  70. Wu Y, Zhu F, Chen W, Zhang M. Effects of transcutaneous electrical nerve stimulation (TENS) in people with knee osteoarthritis: a systematic review and meta-analysis. Clin Rehabil. 2022;36(4):472–85.

    PubMed  Google Scholar 

  71. Johnson MI, Claydon LS, Herbison GP, Jones G, Paley CA. Transcutaneous electrical nerve stimulation (TENS) for fibromyalgia in adults. Cochrane Database Syst Rev. 2017;10(10):CD012172.

    PubMed  Google Scholar 

  72. Cardinali A, Celini D, Chaplik M, Grasso E, Nemec EC. Efficacy of transcutaneous electrical nerve stimulation for postoperative pain, pulmonary function, and opioid consumption following cardiothoracic procedures: a systematic review. Neuromodulation. 2021;24(8):1439–50.

    PubMed  Google Scholar 

  73. Seminowicz D, Mikulis D, Davis K. Cognitive modulation of pain-related brain responses depends on behavioral strategy. Pain. 2004;112(1–2):48–58.

    CAS  PubMed  Google Scholar 

  74. Sulaiman NAZ, Ahmad AI, Zaman MHM. A programmable transcutaneous electrical nerve stimulation device based on arduino and remote control using a smartphone. Int J Adv Technol Eng Explor. 2021;8(75):320.

    Google Scholar 

  75. Zeng D, Hu Y, He Q, Leng B, Wang H, Zou H, Wu W. Study of intelligent bio-feedback therapy system based on transcutaneous electrical nerve stimulation and surface EMG signals. In: 2013 IEEE International Conference on Information and Automation (ICIA); 2013.

  76. Lane E, Latham T. Managing pain using heat and cold therapy. Paediatr Nurs. 2009;21(6):14–8.

    PubMed  Google Scholar 

  77. Brederson J-D, Kym PR, Szallasi A. Targeting TRP channels for pain relief. Eur J Pharmacol. 2013;716(1–3):61–76.

    CAS  PubMed  Google Scholar 

  78. Fahami F, Behmanesh F, Valiani M, Ashouri E. Effect of heat therapy on pain severity in primigravida women. Iran J Nurs Midwifery Res. 2011;16(1):113.

    PubMed  PubMed Central  Google Scholar 

  79. Nadler SF. Nonpharmacologic management of pain. J Osteopath Med. 2004;104(s11):6–12.

    Google Scholar 

  80. Rakel B, Barr JO. Physical modalities in chronic pain management. Nurs Clin. 2003;38(3):477–94.

    Google Scholar 

  81. Thomas D-A, Maslin B, Legler A, Springer E, Asgerally A, Vadivelu N. Role of alternative therapies for chronic pain syndromes. Curr Pain Headache Rep. 2016;20:1–7.

    Google Scholar 

  82. Meeusen R, Lievens P. The use of cryotherapy in sports injuries. Sports Med. 1986;3:398–414.

    CAS  PubMed  Google Scholar 

  83. Douzi W, Dupuy O, Tanneau M, Boucard G, Bouzigon R, Dugué B. 3-min whole body cryotherapy/cryostimulation after training in the evening improves sleep quality in physically active men. Eur J Sport Sci. 2019;19(6):860–7.

    PubMed  Google Scholar 

  84. Mohammadpour A, Najafi S, Bazeli J, Parimoo M. Effect of topical heat therapy on clinical symptoms of peripheral neuropathy in patients with type II diabetes: a clinical trial. HAYAT. 2023;29(1):61–76.

    Google Scholar 

  85. Emine KE, Gulbeyaz C. The effect of salt-water bath in the management of treatment-related peripheral neuropathy in cancer patients receiving taxane and platinum-based treatment. Explore. 2022;18(3):347–56.

    PubMed  Google Scholar 

  86. Henwood P, Ellis JA. Chronic neuropathic pain in spinal cord injury: the patient’s perspective. Pain Res Manag. 2004;9:39–45.

    PubMed  Google Scholar 

  87. Ramsey SD, Spencer AC, Topolski TD, Belza B, Patrick DL. Use of alternative therapies by older adults with osteoarthritis. Arthritis Care Res. 2001;45(3):222–7.

    CAS  Google Scholar 

  88. Denegar CR, Dougherty DR, Friedman JE, Schimizzi ME, Clark JE, Comstock BA, Kraemer WJ. Preferences for heat, cold, or contrast in patients with knee osteoarthritis affect treatment response. Clin Interv Aging. 2010;5:199–206.

    PubMed  PubMed Central  Google Scholar 

  89. McCain GA. Nonmedicinal treatments in primary fibromyalgia. Rheum Dis Clin North Am. 1989;15(1):73–90.

    CAS  PubMed  Google Scholar 

  90. Guillot X, Tordi N, Mourot L, Demougeot C, Dugue B, Prati C, Wendling D. Cryotherapy in inflammatory rheumatic diseases: a systematic review. Expert Rev Clin Immunol. 2014;10(2):281–94.

    CAS  PubMed  Google Scholar 

  91. Pouedras M, Blancheton A, Agneray H, Crenn V, Bellemère P. Effect of cryotherapy on pain and analgesic consumption after wrist or thumb surgery. Hand Surg Rehabil. 2021;40(2):190–3.

    CAS  PubMed  Google Scholar 

  92. Garcia C, Karri J, Zacharias NA, Abd-Elsayed A. Use of cryotherapy for managing chronic pain: an evidence-based narrative. Pain Ther. 2021;10:81–100.

    PubMed  Google Scholar 

  93. El-Tallawy SN, Nalamasu R, Salem GI, LeQuang JAK, Pergolizzi JV, Christo PJ. Management of musculoskeletal pain: an update with emphasis on chronic musculoskeletal pain. Pain Ther. 2021;10:181–209.

    PubMed  PubMed Central  Google Scholar 

  94. Klintberg IH, Larsson ME. Shall we use cryotherapy in the treatment in surgical procedures, in acute pain or injury, or in long term pain or dysfunction?-A systematic review. J Bodyw Mov Ther. 2021;27:368–87.

    PubMed  Google Scholar 

  95. Dehghan M, Farahbod F. The efficacy of thermotherapy and cryotherapy on pain relief in patients with acute low back pain, a clinical trial study. J Clin Diagn Res. 2014;8(9):LC01.

    PubMed  PubMed Central  Google Scholar 

  96. Jo J, Lee SH. Heat therapy for primary dysmenorrhea: a systematic review and meta-analysis of its effects on pain relief and quality of life. Sci Rep. 2018;8(1):16252.

    PubMed  PubMed Central  Google Scholar 

  97. Roberts L. Effects of patterns of pressure application on resting electromyography during massage. Int J Ther Massage Bodywork. 2011;4(1):4.

    PubMed  PubMed Central  Google Scholar 

  98. Field T. Social touch, CT touch and massage therapy: a narrative review. Dev Rev. 2019;51:123–45.

    Google Scholar 

  99. Zhang H, Chen H, Wang H, Li D, Jia B, Tan Z, Zheng B, Weng Z. Effect of Chinese tuina massage therapy on resting state brain functional network of patients with chronic neck pain. J Tradit Chin Med Sci. 2015;2(1):60–8.

    CAS  Google Scholar 

  100. Sunarmi S, Isworo A, Ari D, Sitepu FY, Triredjeki H. The effectiveness of massage therapy on healing of diabetic neuropathy in diabetes mellitus patients. Open Access Maced J Med Sci. 2022;10(G):190–4.

    Google Scholar 

  101. Lopez G, Eng C, Overman M, Ramirez D, Liu W, Beinhorn C, Sumler P, Prinsloo S, Li Y, Chen M. A randomized pilot study of oncology massage to treat chemotherapy-induced peripheral neuropathy. Sci Rep. 2022;12(1):19023.

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Xing X-X, Zheng M-X, Hua X-Y, Ma S-J, Ma Z-Z, Xu J-G. Brain plasticity after peripheral nerve injury treatment with massage therapy based on resting-state functional magnetic resonance imaging. Neural Regen Res. 2021;16(2):388.

    PubMed  Google Scholar 

  103. Field T. Knee osteoarthritis pain in the elderly can be reduced by massage therapy, yoga and tai chi: a review. Complement Ther Clin Pract. 2016;22:87–92.

    PubMed  Google Scholar 

  104. Quinn C, Chandler C, Moraska A. Massage therapy and frequency of chronic tension headaches. Am J Public Health. 2002;92(10):1657–61.

    PubMed  PubMed Central  Google Scholar 

  105. Jackson JL, Mancuso JM, Nickoloff S, Bernstein R, Kay C. Tricyclic and tetracyclic antidepressants for the prevention of frequent episodic or chronic tension-type headache in adults: a systematic review and meta-analysis. J Gen Intern Med. 2017;32:1351–8.

    PubMed  PubMed Central  Google Scholar 

  106. Yang M, Feng Y, Pei H, Deng S, Wang M, Xiao X, Zheng H, Lai Z, Chen J, Li X. Effectiveness of Chinese massage therapy (Tui Na) for chronic low back pain: study protocol for a randomized controlled trial. Trials. 2014;15(1):1–7.

    Google Scholar 

  107. Kamali F, Panahi F, Ebrahimi S, Abbasi L. Comparison between massage and routine physical therapy in women with sub acute and chronic nonspecific low back pain. J Back Musculoskelet Rehabil. 2014;27(4):475–80.

    PubMed  Google Scholar 

  108. Bervoets DC, Luijsterburg PA, Alessie JJ, Buijs MJ, Verhagen AP. Massage therapy has short-term benefits for people with common musculoskeletal disorders compared to no treatment: a systematic review. J Physiother. 2015;61(3):106–16.

    PubMed  Google Scholar 

  109. Kennedy AB, Trilk JL. A standardized, evidence-based massage therapy program for decentralized elite paracyclists: creating the model. Int J Ther Massage Bodywork. 2015;8(3):3.

    PubMed  PubMed Central  Google Scholar 

  110. McGrath JM. Touch and massage in the newborn period: effects on biomarkers and brain development. J Perinat Neonatal Nurs. 2009;23(4):304–6.

    PubMed  Google Scholar 

  111. Field T, Hemandez-Reif M, Hart S, Theakston H, Schanberg S, Kuhn C. Pregnant women benefit from massage therapy. J Psychosom Obstet Gynaecol. 1999;20(1):31–8.

    CAS  PubMed  Google Scholar 

  112. Chen FR, Manzi JE, Mehta N, Gulati A, Jones M. A review of laser therapy and low-intensity ultrasound for chronic pain states. Curr Pain Headache Rep. 2022;26(1):57–63.

    PubMed  Google Scholar 

  113. Aiyer R, Noori SA, Chang KV, Jung B, Rasheed A, Bansal N, Ottestad E, Gulati A. Therapeutic ultrasound for chronic pain management in joints: a systematic review. Pain Med. 2020;21(7):1437–48.

  114. Zhou X-Y, Xu X-M, Wu S-Y, Wang F, Zhang Z-C, Yang Y-L, Li M, Wei X-Z. Low-intensity pulsed ultrasound-induced spinal fusion is coupled with enhanced calcitonin gene-related peptide expression in rat model. Ultrasound Med Biol. 2017;43(7):1486–93.

    PubMed  Google Scholar 

  115. Paoloni M, Tavernese E, Cacchio A, D’orazi V, Ioppolo F, Fini M, Santilli V, Mangone M. Extracorporeal shock wave therapy and ultrasound therapy improve pain and function in patients with carpal tunnel syndrome. A randomized controlled trial. Eur J Phys Rehabil Med. 2015;51(5):521–8.

    CAS  PubMed  Google Scholar 

  116. Chu Y-C, Lim J, Chien A, Chen C-C, Wang J-L. Activation of mechanosensitive ion channels by ultrasound. Ultrasound Med Biol. 2022;48:1981.

    PubMed  Google Scholar 

  117. Matesanz-García L, Schmid AB, Cáceres-Pajuelo JE, Cuenca-Martínez F, Arribas-Romano A, González-Zamorano Y, Goicoechea-García C, Fernández-Carnero J. Effect of physiotherapeutic interventions on biomarkers of neuropathic pain: a systematic review of preclinical literature. J Pain. 2022;23(11):1833–55.

    PubMed  Google Scholar 

  118. Nagata K, Nakamura T, Fujihara S, Tanaka E. Ultrasound modulates the inflammatory response and promotes muscle regeneration in injured muscles. Ann Biomed Eng. 2013;41:1095–105.

    PubMed  Google Scholar 

  119. Lin G, Reed-Maldonado AB, Lin M, Xin Z, Lue TF. Effects and mechanisms of low-intensity pulsed ultrasound for chronic prostatitis and chronic pelvic pain syndrome. Int J Mol Sci. 2016;17(7):1057.

    PubMed  PubMed Central  Google Scholar 

  120. Sanguinetti JL, Hameroff S, Smith EE, Sato T, Daft CM, Tyler WJ, Allen JJ. Transcranial focused ultrasound to the right prefrontal cortex improves mood and alters functional connectivity in humans. Front Hum Neurosci. 2020;14:52.

    PubMed  PubMed Central  Google Scholar 

  121. Nüssel M, Zhao Y, Knorr C, Regensburger M, Stadlbauer A, Buchfelder M, Del Vecchio A, Kinfe T. Deep brain stimulation, stereotactic radiosurgery and high-intensity focused ultrasound targeting the limbic pain matrix: a comprehensive review. Pain Ther. 2022;11(2):459–76.

    PubMed  PubMed Central  Google Scholar 

  122. Freitas LS, Freitas TP, Silveira PC, Rocha LG, Pinho RA, Streck EL. Effect of therapeutic pulsed ultrasound on parameters of oxidative stress in skeletal muscle after injury. Cell Biol Int. 2007;31(5):482–8.

    CAS  PubMed  Google Scholar 

  123. Martins C, Moraes M, Hauck M, Guerreiro L, Rossato D, Varela A Jr, Da Rosa C, Signori L. Effects of cryotherapy combined with therapeutic ultrasound on oxidative stress and tissue damage after musculoskeletal contusion in rats. Physiotherapy. 2016;102(4):377–83.

    CAS  PubMed  Google Scholar 

  124. Noori SA, Rasheed A, Aiyer R, Jung B, Bansal N, Chang K-V, Ottestad E, Gulati A. Therapeutic ultrasound for pain management in chronic low back pain and chronic neck pain: a systematic review. Pain Med. 2020;21(7):1482–93.

    PubMed  Google Scholar 

  125. Tan J-S, Lin C-C, Chen G-S. Vasomodulation of peripheral blood flow by focused ultrasound potentiates improvement of diabetic neuropathy. BMJ Open Diabetes Res Care. 2020;8(1):e001004.

    PubMed  PubMed Central  Google Scholar 

  126. Payne C. Ultrasound for post-herpetic neuralgia. A study to investigate the results of treatment. Physiotherapy. 1984;70(3):96–7.

    CAS  PubMed  Google Scholar 

  127. Wijntjes J, Borchert A, van Alfen N. Nerve ultrasound in traumatic and iatrogenic peripheral nerve injury. Diagnostics. 2020;11(1):30.

    PubMed  PubMed Central  Google Scholar 

  128. Almeida TF, Roizenblatt S, Benedito-Silva AA, Tufik S. The effect of combined therapy (ultrasound and interferential current) on pain and sleep in fibromyalgia. Pain. 2003;104(3):665–72.

    PubMed  Google Scholar 

  129. Kozanoglu E, Basaran S, Guzel R, Guler-Uysal F. Short term efficacy of ibuprofen phonophoresis versus continuous ultrasound therapy in knee osteoarthritis. Swiss Med Wkly. 2003;133(2324):333–8.

    CAS  PubMed  Google Scholar 

  130. Rafanan BS Jr, Valdecañas BF, Lim BP, Malairungsakul A, Tassanawipas W, Shiyi C, Tse LF, Luong TK. Consensus recommendations for managing osteoarthritic pain with topical NSAIDs in Asia-Pacific. Pain Manag. 2018;8(2):115–28.

    PubMed  Google Scholar 

  131. Connelly NR, Malik A, Madabushi L, Gibson C. Use of ultrasound-guided cryotherapy for the management of chronic pain states. J Clin Anesth. 2013;25(8):634–6.

    PubMed  Google Scholar 

  132. Ucar M, Sarp Ü, Koca İ, Eroğlu S, Yetisgin A, Tutoglu A, Boyacı A. Effectiveness of a home exercise program in combination with ultrasound therapy for temporomandibular joint disorders. J Phys Ther Sci. 2014;26(12):1847–9.

    PubMed  PubMed Central  Google Scholar 

  133. Loyola-Sánchez A, Richardson J, MacIntyre N. Efficacy of ultrasound therapy for the management of knee osteoarthritis: a systematic review with meta-analysis. Osteoarthritis Cartilage. 2010;18(9):1117–26.

    PubMed  Google Scholar 

  134. Flynn DM. Chronic musculoskeletal pain: nonpharmacologic noninvasive treatments. Am Fam Physician. 2020;102(8):465–77.

    PubMed  Google Scholar 

  135. Stamm O, Perotti L. Expert requirements for an ultrasound-based wearable using deep learning for exercise feedback in older chronic back pain patients. In: International Conference on Human-Computer Interaction; 2022.

  136. DEO MF, Johnson DS, Demchak T, Tomazoni SS, Leal-Junior EC. Low-intensity LASER and LED (photobiomodulation therapy) for pain control of the most common musculoskeletal conditions. Eur J Phys Rehabil Med. 2022;58(2):282–9.

    Google Scholar 

  137. Eroy-Reveles AA, Leung Y, Beavers CM, Olmstead MM, Mascharak PK. Near-infrared light activated release of nitric oxide from designed photoactive manganese nitrosyls: strategy, design, and potential as NO donors. J Am Chem Soc. 2008;130(13):4447–58.

    CAS  PubMed  Google Scholar 

  138. Tam G. Low power laser therapy and analgesic action. J Clin Laser Med Surg. 1999;17(1):29–33.

    CAS  PubMed  Google Scholar 

  139. Hamblin MR, Demidova TN. Mechanisms of low level light therapy. Mech Low-Light Ther. 2006;6140:614001.

    Google Scholar 

  140. Lang-Illievich K, Winter R, Rumpold-Seitlinger G, Schicho K, Dorn C, Klivinyi C, Bornemann-Cimenti H. The effect of low-level light therapy on capsaicin-induced peripheral and central sensitization in healthy volunteers: a double-blinded, randomized. Sham-Controlled Trial Pain Ther. 2020;9:717–26.

    PubMed  Google Scholar 

  141. Thunshelle C, Hamblin MR. Transcranial Low-level laser (Light) therapy for brain injury. Photomed Laser Surg. 2016;34(12):587–98.

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Chung H, Dai T, Sharma SK, Huang YY, Carroll JD, Hamblin MR. The nuts and bolts of low-level laser (light) therapy. Ann Biomed Eng. 2012;40(2):516–33.

    PubMed  Google Scholar 

  143. Bridges M, Hilliard J, Chui K. Effects of light therapy on osteoarthritis and its sequelae in aging and older adults: a systematic narrative review. Top Geriatr Rehabil. 2020;36(1):11–37.

    Google Scholar 

  144. Chow RT, Barnsley L. Systematic review of the literature of low-level laser therapy (LLLT) in the management of neck pain. Lasers Surg Med. 2005;37(1):46–52.

    PubMed  Google Scholar 

  145. Martin L, Porreca F, Mata EI, Salloum M, Goel V, Gunnala P, Killgore WD, Jain S, Jones-MacFarland FN, Khanna R. Green light exposure improves pain and quality of life in fibromyalgia patients: a preliminary one-way crossover clinical trial. Pain Med. 2021;22(1):118–30.

    PubMed  Google Scholar 

  146. Wang J-Y, Huang Z-Q, Deng H-P, Zhao L, Deng H-Y, Liu J-P, Shen X-Y, Cheng K. Low level light therapy/photobiomodulation for diabetic peripheral neuropathy: protocol of a systematic review and meta-analysis. BMJ Open. 2022;12(9):e059476.

    PubMed  PubMed Central  Google Scholar 

  147. Lee JM, Look RM, Turner C, Gardiner SK, Wagie T, Douglas J, et al. Low-level laser therapy for chemotherapy-induced peripheral neuropathy. Am Soc Clin Oncol. 2012;30:9019.

    Google Scholar 

  148. Leskowitz E. Energy medicine perspectives on phantom-limb pain. Altern Complement Ther. 2009;15(2):59–63.

    Google Scholar 

  149. Mukhtar R, Fazal MU, Saleem Ma, Saleem S. Role of low-level laser therapy in post-herpetic neuralgia: a pilot study. Lasers Med Sci. 2020;35:1759–64.

    PubMed  Google Scholar 

  150. Zlatkovic-Svenda MI, Leitner C, Lazovic B, Petrovic DM. Complex regional pain syndrome (Sudeck atrophy) prevention possibility and accelerated recovery in patients with distal radius at the typical site fracture using polarized, polychromatic light therapy. Photobiomodul Photomed Laser Surg. 2019;37(4):233–9.

    PubMed  Google Scholar 

  151. Hamblin MR. Can osteoarthritis be treated with light? Arthritis Res Ther. 2013;15:120.

    PubMed  PubMed Central  Google Scholar 

  152. Liu D, Kim B, Fu W, Zhu S, Kang J, Kim O, et al. Upturn strategies for arachidonic acid-induced MC3T3-E1—625 nm irradiation in combination with NSAIDs: dissipating inflammation and promoting healing. Photonics. 2023;10(5):535.

    CAS  Google Scholar 

  153. Wickenheisser VA, Zywot EM, Rabjohns EM, Lee HH, Lawrence DS, Tarrant TK. Laser light therapy in inflammatory, musculoskeletal, and autoimmune disease. Curr Allergy Asthma Rep. 2019;19:1–15.

    CAS  Google Scholar 

  154. Short S, Pace G, Birnbaum C. Nonpharmacologic techniques to assist in pediatric pain management. Clin Pediatr Emerg Med. 2017;18(4):256–60.

    Google Scholar 

  155. Lam RW, Tam EM. A clinician’s guide to using light therapy. New York: Cambridge University Press; 2009.

    Google Scholar 

  156. Knoerl R, Lavoie Smith EM, Weisberg J. Chronic pain and cognitive behavioral therapy: an integrative review. West J Nurs Res. 2016;38(5):596–628.

    PubMed  Google Scholar 

  157. Heffner KL, France CR, Ashrafioun L, Quiñones M, Walsh P, Maloney MD, Giordano BD, Pigeon WR. Clinical pain-related outcomes and inflammatory cytokine response to pain following insomnia improvement in adults with knee osteoarthritis. Clin J Pain. 2018;34(12):1133–40.

    PubMed  PubMed Central  Google Scholar 

  158. Yoshino A, Okamoto Y, Okada G, Takamura M, Ichikawa N, Shibasaki C, Yokoyama S, Doi M, Jinnin R, Yamashita H, et al. Changes in resting-state brain networks after cognitive-behavioral therapy for chronic pain. Psychol Med. 2018;48(7):1148–56.

    CAS  PubMed  Google Scholar 

  159. Mariano TY, Urman RD, Hutchison CA, Jamison RN, Edwards RR. Cognitive Behavioral Therapy (CBT) for subacute low back pain: a systematic review. Curr Pain Headache Rep. 2018;22(3):15.

    PubMed  Google Scholar 

  160. Clauw DJ. Fibromyalgia: a clinical review. JAMA. 2014;311(15):1547–55.

    PubMed  Google Scholar 

  161. Higgins DM, Heapy AA, Buta E, LaChappelle KM, Serowik KL, Czlapinski R, Kerns RD. A randomized controlled trial of cognitive behavioral therapy compared with diabetes education for diabetic peripheral neuropathic pain. J Health Psychol. 2022;27(3):649–62.

    PubMed  Google Scholar 

  162. Shen B, Li Y, Du X, Chen H, Xu Y, Li H, Xu GY. Effects of cognitive behavioral therapy for patients with rheumatoid arthritis: a systematic review and meta-analysis. Psychol Health Med. 2020;25(10):1179–91.

    PubMed  Google Scholar 

  163. Kinsinger SW. Cognitive-behavioral therapy for patients with irritable bowel syndrome: current insights. Psychol Res Behav Manag. 2017;10:231–7.

    PubMed  PubMed Central  Google Scholar 

  164. Randhawa K, Bohay R, Côté P, van der Velde G, Sutton D, Wong JJ, Yu H, Southerst D, Varatharajan S, Mior S, et al. The effectiveness of noninvasive interventions for temporomandibular disorders: a systematic review by the Ontario Protocol for Traffic Injury Management (OPTIMa) collaboration. Clin J Pain. 2016;32(3):260–78.

    PubMed  Google Scholar 

  165. Levy RL, Langer SL, Walker LS, Romano JM, Christie DL, Youssef N, DuPen MM, Feld AD, Ballard SA, Welsh EM, et al. Cognitive-behavioral therapy for children with functional abdominal pain and their parents decreases pain and other symptoms. Am J Gastroenterol. 2010;105(4):946–56.

    PubMed  PubMed Central  Google Scholar 

  166. Urits I, Callan J, Moore WC, Fuller MC, Renschler JS, Fisher P, Jung JW, Hasoon J, Eskander J, Kaye AD, et al. Cognitive behavioral therapy for the treatment of chronic pelvic pain. Best Pract Res Clin Anaesthesiol. 2020;34(3):409–26.

    PubMed  Google Scholar 

  167. Maindet C, Burnod A, Minello C, George B, Allano G, Lemaire A. Strategies of complementary and integrative therapies in cancer-related pain-attaining exhaustive cancer pain management. Support Care Cancer. 2019;27(8):3119–32.

    PubMed  Google Scholar 

  168. Khan M, Akhter S, Soomro RR, Ali SS. The effectiveness of Cognitive Behavioral Therapy (CBT) with general exercises versus general exercises alone in the management of chronic low back pain. Pak J Pharm Sci. 2014;27(4 Suppl):1113–6.

    PubMed  Google Scholar 

  169. Turner JA, Mancl L, Aaron LA. Short- and long-term efficacy of brief cognitive-behavioral therapy for patients with chronic temporomandibular disorder pain: a randomized, controlled trial. Pain. 2006;121(3):181–94.

    PubMed  Google Scholar 

  170. Alashram AR, Annino G, Padua E, Romagnoli C, Mercuri NB. Cognitive rehabilitation post traumatic brain injury: a systematic review for emerging use of virtual reality technology. J Clin Neurosci. 2019;66:209–19.

    PubMed  Google Scholar 

  171. Rubin R. Virtual reality device is authorized to relieve back pain. JAMA. 2021;326(23):2354.

    CAS  PubMed  Google Scholar 

  172. Evers AW, Kraaimaat FW, van Riel PL, de Jong AJ. Tailored cognitive-behavioral therapy in early rheumatoid arthritis for patients at risk: a randomized controlled trial. Pain. 2002;100(1–2):141–53.

    PubMed  Google Scholar 

  173. Jensen MP. Hypnosis for chronic pain management: a new hope. Pain. 2009;146(3):235–7.

    PubMed  Google Scholar 

  174. Jensen M, Patterson DR. Hypnotic treatment of chronic pain. J Behav Med. 2006;29(1):95–124.

    PubMed  Google Scholar 

  175. Derbyshire SW, Whalley MG, Oakley DA. Fibromyalgia pain and its modulation by hypnotic and non-hypnotic suggestion: an fMRI analysis. EurJ Pain. 2009;13(5):542–50.

    Google Scholar 

  176. Del Casale A, Ferracuti S, Rapinesi C, Serata D, Caltagirone SS, Savoja V, Piacentino D, Callovini G, Manfredi G, Sani G. Pain perception and hypnosis: findings from recent functional neuroimaging studies. Int J Clin Exp Hypn. 2015;63(2):144–70.

    PubMed  Google Scholar 

  177. Liu YY, Wang TX, Zhou JC, Qu WM, Huang ZL. Dopamine D(1) and D(2) receptors mediate analgesic and hypnotic effects of l-tetrahydropalmatine in a mouse neuropathic pain model. Psychopharmacology. 2019;236(11):3169–82.

    CAS  PubMed  Google Scholar 

  178. Kupers R, Faymonville ME, Laureys S. The cognitive modulation of pain: hypnosis- and placebo-induced analgesia. Prog Brain Res. 2005;150:251–69.

    PubMed  Google Scholar 

  179. Császár N, Scholkmann F, Bókkon I. Implications on hypnotherapy: neuroplasticity, epigenetics and pain. Neurosci Biobehav Rev. 2021;131:755–64.

    PubMed  Google Scholar 

  180. Presciuttini S, Gialluisi A, Barbuti S, Curcio M, Scatena F, Carli G, Santarcangelo EL. Hypnotizability and Catechol-O-Methyltransferase (COMT) polymorphysms in Italians. Front Hum Neurosci. 2014;7:929.

    PubMed  PubMed Central  Google Scholar 

  181. Tan G, Rintala DH, Jensen MP, Fukui T, Smith D, Williams W. A randomized controlled trial of hypnosis compared with biofeedback for adults with chronic low back pain. Eur J Pain. 2015;19(2):271–80.

    CAS  PubMed  Google Scholar 

  182. Horton-Hausknecht JR, Mitzdorf U, Melchart D. The effect of hypnosis therapy on the symptoms and disease activity in rheumatoid arthritis. Psychol Health. 2000;14(6):1089–104.

    CAS  PubMed  Google Scholar 

  183. Aravena V, García FE, Téllez A, Arias PR. Hypnotic intervention in people with fibromyalgia: a randomized controlled trial. Am J Clin Hypn. 2020;63(1):49–61.

    PubMed  Google Scholar 

  184. Flynn N. Effect of an online hypnosis intervention in reducing migraine symptoms: a randomized controlled trial. Int J Clin Exp Hypn. 2019;67(3):313–35.

    PubMed  Google Scholar 

  185. Sine H, Achbani A, Filali K. The effect of hypnosis on the intensity of pain and anxiety in cancer patients: a systematic review of controlled experimental trials. Cancer Invest. 2022;40(3):235–53.

    PubMed  Google Scholar 

  186. Rousseaux F, Faymonville ME, Nyssen AS, Dardenne N, Ledoux D, Massion PB, Vanhaudenhuyse A. Can hypnosis and virtual reality reduce anxiety, pain and fatigue among patients who undergo cardiac surgery: a randomised controlled trial. Trials. 2020;21(1):330.

    PubMed  PubMed Central  Google Scholar 

  187. Langlois P, Perrochon A, David R, Rainville P, Wood C, Vanhaudenhuyse A, Pageaux B, Ounajim A, Lavallière M, Debarnot U. Hypnosis to manage musculoskeletal and neuropathic chronic pain: a systematic review and meta-analysis. Neurosci Biobehav Rev. 2022;135:104591.

    PubMed  Google Scholar 

  188. Joo Y, Kim EK, Song HG, Jung H, Park H, Moon JY. Effectiveness of virtual reality immersion on procedure-related pain and anxiety in outpatient pain clinic: an exploratory randomized controlled trial. Korean J Pain. 2021;34(3):304–14.

    PubMed  PubMed Central  Google Scholar 

  189. Moujaes FF, Rieser NM, Phillips C, de Matos NMP, Brügger M, Dürler P, et al. Comparing neural correlates of consciousness: from psychedelics to hypnosis and meditation. Biol Psychiatry Cogn Neurosci Neuroimaging. 2023. https://doi.org/10.1016/j.bpsc.2023.07.003. Online first.

  190. Pardos-Gascón EM, Narambuena L, Leal-Costa C, van der Hofstadt-Román CJ. Differential efficacy between cognitive-behavioral therapy and mindfulness-based therapies for chronic pain: systematic review. Int J Clin Health Psychol. 2021;21(1):100197.

    PubMed  Google Scholar 

  191. Pei JH, Ma T, Nan RL, Chen HX, Zhang YB, Gou L, Dou XM. Mindfulness-based cognitive therapy for treating chronic pain a systematic review and meta-analysis. Psychol Health Med. 2021;26(3):333–46.

    PubMed  Google Scholar 

  192. Zeidan F, Baumgartner JN, Coghill RC. The neural mechanisms of mindfulness-based pain relief: a functional magnetic resonance imaging-based review and primer. Pain Rep. 2019;4(4):e759.

    PubMed  PubMed Central  Google Scholar 

  193. Chiesa A, Brambilla P, Serretti A. Functional neural correlates of mindfulness meditations in comparison with psychotherapy, pharmacotherapy and placebo effect. Is there a link? Acta Neuropsychiatr. 2010;22(3):104–17.

    PubMed  Google Scholar 

  194. Gu YQ, Zhu Y. Underlying mechanisms of mindfulness meditation: genomics, circuits, and networks. World J Psychiatry. 2022;12(9):1141–9.

    PubMed  PubMed Central  Google Scholar 

  195. Marinovic DA, Hunter RL. Examining the interrelationships between mindfulness-based interventions, depression, inflammation, and cancer survival. CA Cancer J Clin. 2022;72(5):490–502.

    PubMed  Google Scholar 

  196. Riegner G, Posey G, Oliva V, Jung Y, Mobley W, Zeidan F. Disentangling self from pain: mindfulness meditation-induced pain relief is driven by thalamic-default mode network decoupling. Pain. 2023;164(2):280–91.

    PubMed  Google Scholar 

  197. Leça S, Tavares I. Research in mindfulness interventions for patients with fibromyalgia: a critical review. Front Integr Neurosci. 2022;16:920271.

    PubMed  PubMed Central  Google Scholar 

  198. Zhou B, Wang G, Hong Y, Xu S, Wang J, Yu H, Liu Y, Yu L. Mindfulness interventions for rheumatoid arthritis: a systematic review and meta-analysis. Complement Ther Clin Pract. 2020;39:101088.

    PubMed  Google Scholar 

  199. Faedda N, Natalucci G, Baglioni V, Giannotti F, Cerutti R, Guidetti V. Behavioral therapies in headache: focus on mindfulness and cognitive behavioral therapy in children and adolescents. Expert Rev Neurother. 2019;19(12):1219–28.

    CAS  PubMed  Google Scholar 

  200. Hussain N, Said ASA. Mindfulness-based meditation versus progressive relaxation meditation: impact on chronic pain in older female patients with diabetic neuropathy. J Evid Based Integr Med. 2019;24:2515690x19876599.

    PubMed  PubMed Central  Google Scholar 

  201. Ngamkham S, Holden JE, Smith EL. A systematic review: mindfulness intervention for cancer-related pain. Asia Pac J Oncol Nurs. 2019;6(2):161–9.

    PubMed  PubMed Central  Google Scholar 

  202. Dowsey M, Castle D, Knowles S, Monshat K, Salzberg M, Nelson E, Dunin A, Dunin J, Spelman T, Choong P. The effect of mindfulness training prior to total joint arthroplasty on post-operative pain and physical function: a randomised controlled trial. Complement Ther Med. 2019;46:195–201.

    PubMed  Google Scholar 

  203. Naliboff BD, Smith SR, Serpa JG, Laird KT, Stains J, Connolly LS, Labus JS, Tillisch K. Mindfulness-based stress reduction improves irritable bowel syndrome (IBS) symptoms via specific aspects of mindfulness. Neurogastroenterol Motil. 2020;32(9):e13828.

    PubMed  Google Scholar 

  204. Bittelbrunn CC, de Fraga R, Martins C, Romano R, Massaneiro T, Mello GVP, Canciglieri M. Pelvic floor physical therapy and mindfulness: approaches for chronic pelvic pain in women-a systematic review and meta-analysis. Arch Gynecol Obstet. 2023;307(3):663–72.

    PubMed  Google Scholar 

  205. Penlington C, Bowes C, Taylor G, Otemade AA, Waterhouse P, Durham J, Ohrbach R. Psychological therapies for temporomandibular disorders (TMDs). Cochrane Database Syst Rev. 2022;8(8):Cd013515.

    PubMed  Google Scholar 

  206. Seng EK, Singer AB, Metts C, Grinberg AS, Patel ZS, Marzouk M, Rosenberg L, Day M, Minen MT, Lipton RB, et al. Does Mindfulness-based cognitive therapy for migraine reduce migraine-related disability in people with episodic and chronic migraine? A phase 2b pilot randomized clinical trial. Headache. 2019;59(9):1448–67.

    PubMed  PubMed Central  Google Scholar 

  207. Jones DR, Lehman BJ, Noriega A, Dinnel DL. The effects of a short-term mindfulness meditation intervention on coping flexibility. Anxiety Stress Cop. 2019;32(4):347–61.

    Google Scholar 

  208. Huberty J, Green J, Glissmann C, Larkey L, Puzia M, Lee C. Efficacy of the mindfulness meditation mobile app “Calm” to reduce stress among college students: randomized controlled trial. JMIR Mhealth Uhealth. 2019;7(6):e14273.

    PubMed  PubMed Central  Google Scholar 

  209. Matis J, Svetlak M, Slezackova A, Svoboda M, Šumec R. Mindfulness-based programs for patients with cancer via eHealth and mobile health: systematic review and synthesis of quantitative research. J Med Internet Res. 2020;22(11):e20709.

    PubMed  PubMed Central  Google Scholar 

  210. Wolters F, Peerdeman KJ, Evers AWM. Placebo and nocebo effects across symptoms: from pain to fatigue, dyspnea, nausea, and itch. Front Psychiatr. 2019;10:470.

    Google Scholar 

  211. Egorova N, Benedetti F, Gollub RL, Kong J. Between placebo and nocebo: Response to control treatment is mediated by amygdala activity and connectivity. Eur J Pain. 2020;24(3):580–92.

    PubMed  Google Scholar 

  212. Colloca L, Grillon C. Understanding placebo and nocebo responses for pain management. Curr Pain Headache Rep. 2014;18(6):419.

    PubMed  PubMed Central  Google Scholar 

  213. Colloca L, Wang Y, Martinez PE, Chang YC, Ryan KA, Hodgkinson C, Goldman D, Dorsey SG. OPRM1 rs1799971, COMT rs4680, and FAAH rs324420 genes interact with placebo procedures to induce hypoalgesia. Pain. 2019;160(8):1824–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Meissner K. Placebo, nocebo: Believing in the field of medicine. Front Pain Res (Lausanne). 2022;3:972169.

    PubMed  Google Scholar 

  215. Kaptchuk TJ, Hemond CC, Miller FG. Placebos in chronic pain: evidence, theory, ethics, and use in clinical practice. BMJ. 2020;370:m1668.

    PubMed  Google Scholar 

  216. Zhang W. The powerful placebo effect in osteoarthritis. Clin Exp Rheumatol. 2019;37 Suppl 120(5):118–23.

    PubMed  Google Scholar 

  217. Breivik H. Advances in treatment of neuropathic pain. Eur J Pain. 2002;6 Suppl B:v.

    PubMed  Google Scholar 

  218. Migliorini F, Maffulli N, Eschweiler J, Betsch M, Tingart M, Colarossi G. Placebo effect in pharmacological management of fibromyalgia: a meta-analysis. Br Med Bull. 2021;139(1):73–85.

    CAS  PubMed  Google Scholar 

  219. Enck P, Klosterhalfen S. Placebos and the placebo effect in drug trials. Handb Exp Pharmacol. 2019;260:399–431.

    CAS  PubMed  Google Scholar 

  220. Elsenbruch S, Roderigo T, Enck P, Benson S. Can a brief relaxation exercise modulate placebo or nocebo effects in a visceral pain model? Front Psychiatry. 2019;10:144.

    PubMed  PubMed Central  Google Scholar 

  221. Forbes RB, McCarron M, Cardwell CR. Efficacy and contextual (Placebo) effects of CGRP antibodies for migraine: systematic review and meta-analysis. Headache. 2020;60(8):1542–57.

    PubMed  Google Scholar 

  222. Yin M, Muhammed SA, Wang Y, Colloca L. Yoga and massage are associated with small experimental placebo effects in chronic orofacial pain. Eur J Pain. 2023;27(7):816–30.

    PubMed  Google Scholar 

  223. Vase L, Wartolowska K. Pain, placebo, and test of treatment efficacy: a narrative review. Br J Anaesth. 2019;123(2):e254–62.

    PubMed  PubMed Central  Google Scholar 

  224. Oyamaguchi A, Hanamoto H, Tanaka Y, Takahashi S, Niwa H. Evaluation of the effects of manual acupuncture and electroacupuncture at LI4 and LI11 on perception thresholds: a prospective crossover trial. Acupunct Med. 2023;41(4):206–14.

    PubMed  Google Scholar 

  225. Shi Y, Dai Q, Ji B, Huang L, Zhuang X, Mo Y, Wang J. Electroacupuncture pretreatment prevents cognitive impairment induced by cerebral ischemia-reperfusion via adenosine A1 receptors in rats. Front Aging Neurosci. 2021;13:680706.

    CAS  PubMed  PubMed Central  Google Scholar 

  226. Xu M, Fei Y, He Q, Fu J, Zhu J, Tao J, Ni C, Xu C, Zhou Q, Yao M, et al. Electroacupuncture attenuates cancer-induced bone pain via NF-κB/CXCL12 signaling in midbrain periaqueductal gray. ACS Chem Neurosci. 2021;12(18):3323–34.

    CAS  PubMed  Google Scholar 

  227. Gao J, Zhou N, Lu M, Wang Q, Zhao C, Wang J, Zhou M, Xu Y. Effects of electroacupuncture on urinary metabolome and microbiota in presenilin1/2 conditional double knockout mice. Front Microbiol. 2022;13:1047121.

    PubMed  Google Scholar 

  228. Wu X, Zhou Y, Chen G, Zheng C, Dong H, Xiong F, Zhang M, Huang G, Xu X. Effect of electroacupuncture with different current intensities on the serum metabolomics of functional constipation. Evid Based Complement Alternat Med. 2023;2023:9693390.

    PubMed  PubMed Central  Google Scholar 

  229. Tan LH, Li KG, Wu YY, Guo MW, Lan Y, Wang S, Zhu WL, Ren XX. Effect of electroacupuncture at different acupoints on the expression of NMDA receptors in ACC and colon in IBS rats. Evid Based Complement Alternat Med. 2019;2019:4213928.

    PubMed  PubMed Central  Google Scholar 

  230. Torres SF, de Macedo ACB, Sakai RY, Bressan GCS, Dos Santos MBR, Marques AP. Effect of different frequencies of electroacupuncture on chronic low back pain in older adults: a triple-blind, placebo-controlled, randomized clinical trial. Pain physician. 2023;26(2):161–73.

    PubMed  Google Scholar 

  231. Li P, Zhang Y, Li F, Cai F, Xiao B, Yang H. The efficacy of electroacupuncture in the treatment of knee osteoarthritis: a systematic review and meta-analysis. Adv Biol. 2023:e2200304. https://doi.org/10.1002/adbi.202200304.

  232. Han QQ, Fu Y, Le JM, Ma YJ, Wei XD, Ji HL, Jiang H, Gao Y, Wu H. The therapeutic effects of acupuncture and electroacupuncture on cancer-related symptoms and side-effects. J Cancer. 2021;12(23):7003–9.

    PubMed  PubMed Central  Google Scholar 

  233. Xue X, Liu X, Pan S, Li J, Wang S, Yuan H, Liu Y, Yue Z. Electroacupuncture treatment of primary dysmenorrhea: a randomized, participant-blinded, sham-controlled clinical trial protocol. PLoS One. 2023;18(5):e0282541.

    CAS  PubMed  PubMed Central  Google Scholar 

  234. Li X, Dai Q, Shi Z, Chen H, Hu Y, Wang X, Zhang X, Tian G. Clinical efficacy and safety of electroacupuncture in migraine treatment: a systematic review and network meta-analysis. Am J Chin Med. 2019;47(8):1755–80.

    PubMed  Google Scholar 

  235. Hu J, Wang X, Jia S, Kong L, Wang Y, Xin X, Hu Y, Chen X. Acupuncture and related therapies for tension-type headache: a systematic review and network meta-analysis. Front Neurol. 2023;14:1194441.

    PubMed  PubMed Central  Google Scholar 

  236. Chen KB, Huang Y, Jin XL, Chen GF. Electroacupuncture or transcutaneous electroacupuncture for postoperative ileus after abdominal surgery: a systematic review and meta-analysis. Int J Surg. 2019;70:93–101.

    PubMed  Google Scholar 

  237. Hwang MS, Lee HY, Choi TY, Lee JH, Ko YS, Jo DC, Do K, Lee JH, Park TY. A systematic review and meta-analysis of the efficacy of acupuncture and electroacupuncture against chemotherapy-induced peripheral neuropathy. Medicine. 2020;99(17):e19837.

    PubMed  PubMed Central  Google Scholar 

  238. Jiang B, Zhong X, Fang J, Zhang A, Wang DW, Liang Y, Fang J, Chen F, Du J. Electroacupuncture attenuates morphine tolerance in rats with bone cancer pain by inhibiting PI3K/Akt/JNK1/2 signaling pathway in the spinal dorsal horn. Integr Cancer Ther. 2021;20:1534735421995237.

    CAS  PubMed  PubMed Central  Google Scholar 

  239. Zhang YY, Chen QL, Wang Q, Ding SS, Li SN, Chen SJ, Lin XJ, Li CD, Asakawa T. Role of parameter setting in electroacupuncture: current scenario and future prospects. Chin J Integr Med. 2022;28(10):953–60.

    PubMed  Google Scholar 

  240. Park J, Krause-Parello CA, Barnes CM. A narrative review of movement-based mind-body interventions: effects of yoga, tai chi, and qigong for back pain patients. Holist Nurs Pract. 2020;34(1):3–23.

    PubMed  Google Scholar 

  241. Ferro Moura Franco K, Lenoir D, Dos Santos Franco YR, Jandre Reis FJ, Nunes Cabral CM, Meeus M. Prescription of exercises for the treatment of chronic pain along the continuum of nociplastic pain: a systematic review with meta-analysis. Eur J Pain. 2021;25(1):51–70.

    PubMed  Google Scholar 

  242. Oh B, Bae K, Lamoury G, Eade T, Boyle F, Corless B, Clarke S, Yeung A, Rosenthal D, Schapira L, et al. The effects of tai chi and qigong on immune responses: a systematic review and meta-analysis. Medicines (Basel). 2020;7(7):39.

    CAS  PubMed  Google Scholar 

  243. Mullapudi T, Debnath M, Govindaraj R, Raj P, Banerjee M, Varambally S. Effects of a six-month yoga intervention on the immune-inflammatory pathway in antipsychotic-stabilized schizophrenia patients: a randomized controlled trial. Asian J Psychiatr. 2023;86:103636.

    PubMed  Google Scholar 

  244. Rivest-Gadbois E, Boudrias M-H. What are the known effects of yoga on the brain in relation to motor performances, body awareness and pain? A narrative review. Complement Ther Med. 2019;44:129–42.

    PubMed  Google Scholar 

  245. McCarberg B, Peppin J. Pain pathways and nervous system plasticity: learning and memory in pain. Pain Med. 2019;20(12):2421–37.

    PubMed  Google Scholar 

  246. van Aalst J, Ceccarini J, Demyttenaere K, Sunaert S, Van Laere K. What has neuroimaging taught us on the neurobiology of yoga? A review. Front Integr Neurosci. 2020;14:34.

    PubMed  PubMed Central  Google Scholar 

  247. Shen CL, Watkins BA, Kahathuduwa C, Chyu MC, Zabet-Moghaddam M, Elmassry MM, Luk HY, Brismée JM, Knox A, Lee J, et al. Tai chi improves brain functional connectivity and plasma lysophosphatidylcholines in postmenopausal women with knee osteoarthritis: an exploratory pilot study. Front Med (Lausanne). 2021;8:775344.

    PubMed  Google Scholar 

  248. Schranner D, Kastenmüller G, Schönfelder M, Römisch-Margl W, Wackerhage H. Metabolite concentration changes in humans after a bout of exercise: a systematic review of exercise metabolomics studies. Sports Med Open. 2020;6(1):11.

    PubMed  PubMed Central  Google Scholar 

  249. Soplinska A, Zareba L, Wicik Z, Eyileten C, Jakubik D, Siller-Matula JM, De Rosa S, Malek LA, Postula M. MicroRNAs as biomarkers of systemic changes in response to endurance exercise-a comprehensive review. Diagnostics (Basel). 2020;10(10):813.

    CAS  PubMed  Google Scholar 

  250. Qin J, Zhang Y, Wu L, He Z, Huang J, Tao J, Chen L. Effect of Tai Chi alone or as additional therapy on low back pain: Systematic review and meta-analysis of randomized controlled trials. Medicine. 2019;98(37):e17099.

    PubMed  PubMed Central  Google Scholar 

  251. Hu L, Wang Y, Liu X, Ji X, Ma Y, Man S, Hu Z, Cheng J, Huang F. Tai Chi exercise can ameliorate physical and mental health of patients with knee osteoarthritis: systematic review and meta-analysis. Clin Rehabil. 2021;35(1):64–79.

    PubMed  Google Scholar 

  252. Cheng CA, Chiu YW, Wu D, Kuan YC, Chen SN, Tam KW. Effectiveness of Tai Chi on fibromyalgia patients: a meta-analysis of randomized controlled trials. Complement Ther Med. 2019;46:1–8.

    PubMed  Google Scholar 

  253. Wang S, Tian L, Ma T, Wong YT, Yan LJ, Gao Y, Zhang D, Hui SS, Xie YJ. Effectiveness of tai chi on blood pressure, stress, fatigue, and sleep quality among Chinese women with episodic migraine: a randomised controlled trial. Evid Based Complement Alternat Med. 2022;2022:2089139.

    PubMed  PubMed Central  Google Scholar 

  254. Kandah M, Wilson C, Pilitsis JG. Role of integrative health on neuropathic pain. Curr Pain Headache Rep. 2023;27(4):49–55.

    PubMed  Google Scholar 

  255. Wang Q, Mi S, Yu Z, Li Q, Lei J. Opening a window on attention: adjuvant therapies for inflammatory bowel disease. Can J Gastroenterol Hepatol. 2020;2020:7397523.

    PubMed  PubMed Central  Google Scholar 

  256. Lazaridou A, Koulouris A, Dorado K, Chai P, Edwards RR, Schreiber KL. The impact of a daily yoga program for women with fibromyalgia. Int J Yoga. 2019;12(3):206–17.

    PubMed  PubMed Central  Google Scholar 

  257. Anheyer D, Klose P, Lauche R, Saha FJ, Cramer H. Yoga for treating headaches: a systematic review and meta-analysis. J Gen Intern Med. 2020;35(3):846–54.

    PubMed  Google Scholar 

  258. Mao W, Wang T, Sun M, Zhang F, Li L. Effects of tai chi on postural control in people with peripheral neuropathy: a systematic review with meta-analysis. Healthcare (Basel). 2023;11(11):1559.

    PubMed  Google Scholar 

  259. Ye J, Zheng Q, Zou L, Yu Q, Veronese N, Grabovac I, Stefanac S, Tzeng HM, Yu JJ. Mindful exercise (baduanjin) as an adjuvant treatment for older adults (60 years old and over) of knee osteoarthritis: a randomized controlled trial. Evid Based Complement Alternat Med. 2020;2020:9869161.

    PubMed  PubMed Central  Google Scholar 

  260. Yu N, Huang YT. Important factors affecting user experience design and satisfaction of a mobile health app-a case study of daily yoga app. Int J Environ Res Public Health. 2020;17(19):6967.

    PubMed  PubMed Central  Google Scholar 

  261. Lin ZW, Huang ST, Xu N, Cao H, Chen LW, Chen Q. Effect of music therapy on the chronic pain and midterm quality of life of patients after mechanical valve replacement. Ann Thorac Cardiovasc Surg. 2020;26(4):196–201.

    PubMed  PubMed Central  Google Scholar 

  262. Lin ZW, Liu JF, Xie WP, Chen Q, Cao H. The effect of music therapy on chronic pain, quality of life and quality of sleep in adolescents after transthoracic occlusion of ventricular septal defect. Heart Surg Forum. 2021;24(2):E305–10.

    PubMed  Google Scholar 

  263. Antioch I, Furuta T, Uchikawa R, Okumura M, Otogoto J, Kondo E, Sogawa N, Ciobica A, Tomida M. Favorite music mediates pain-related responses in the anterior cingulate cortex and skin pain thresholds. J Pain Res. 2020;13:2729–37.

    PubMed  PubMed Central  Google Scholar 

  264. Sihvonen AJ, Pitkäniemi A, Särkämö T, Soinila S. Isn’t there room for music in chronic pain management? J Pain. 2022;23(7):1143–50.

    PubMed  Google Scholar 

  265. Aktaş YY, Karabulut N. The use of cold therapy, music therapy and lidocaine spray for reducing pain and anxiety following chest tube removal. Complement Ther Clin Pract. 2019;34:179–84.

    PubMed  Google Scholar 

  266. Lin CL, Hwang SL, Jiang P, Hsiung NH. Effect of music therapy on pain after orthopedic surgery-a systematic review and meta-analysis. Pain Pract. 2020;20(4):422–36.

    PubMed  Google Scholar 

  267. Parlongue G, Cerdan EV, Koenig J, Williams DP. Smartphone based music intervention in the treatment of episodic migraine headaches - a pilot trial. Complement Ther Med. 2021;63:102779.

    PubMed  Google Scholar 

  268. Honzel E, Murthi S, Brawn-Cinani B, Colloca G, Kier C, Varshney A, Colloca L. Virtual reality, music, and pain: developing the premise for an interdisciplinary approach to pain management. Pain. 2019;160(9):1909–19.

    PubMed  PubMed Central  Google Scholar 

  269. Kemper KJ, Danhauer SC. Music as therapy. South Med J. 2005;98(3):282–8.

    PubMed  Google Scholar 

  270. Noma N, Watanabe Y, Shimada A, Usuda S, Iida T, Shimada A, Tanaka Y, Oono Y, Sasaki K. Effects of cognitive behavioral therapy on orofacial pain conditions. J Oral Sci. 2020;63(1):4–7.

    PubMed  Google Scholar 

  271. Yang T, Wang S, Wang R, Wei Y, Kang Y, Liu Y, Zhang C. Effectiveness of five-element music therapy in cancer patients: a systematic review and meta-analysis. Complement Ther Clin Pract. 2021;44:101416.

    PubMed  Google Scholar 

  272. de Melo LA, Bezerra de Medeiros AK, Campos M, Bastos Machado de Resende CM, Barbosa GAS, de Almeida EO. Manual therapy in the treatment of myofascial pain related to temporomandibular disorders: a systematic review. J Oral Facial Pain Headache. 2020;34(2):141–8.

    PubMed  Google Scholar 

  273. Garrido-Ardila EM, Santos-Domínguez M, Rodríguez-Mansilla J, Torres-Piles ST, Rodríguez-Domínguez MT, González-Sánchez B, Jiménez-Palomares M. A systematic review of the effectiveness of virtual reality-based interventions on pain and range of joint movement associated with burn injuries. J Pers Med. 2022;12(8):1269.

    PubMed  PubMed Central  Google Scholar 

  274. Cerritelli F, Chiera M, Abbro M, Megale V, Esteves J, Gallace A, Manzotti A. The challenges and perspectives of the integration between virtual and augmented reality and manual therapies. Front Neurol. 2021;12:700211.

    PubMed  PubMed Central  Google Scholar 

  275. Abdullahi A, Truijen S, Saeys W. Neurobiology of recovery of motor function after stroke: the central nervous system biomarker effects of constraint-induced movement therapy. Neural Plast. 2020;2020:9484298.

    PubMed  PubMed Central  Google Scholar 

  276. van Griensven H, Schmid A, Trendafilova T, Low M. Central sensitization in musculoskeletal pain: lost in translation? J Orthop Sports Phys Ther. 2020;50(11):592–6.

    PubMed  Google Scholar 

  277. Cerritelli F, Chiacchiaretta P, Gambi F, Perrucci MG, Barassi G, Visciano C, Bellomo RG, Saggini R, Ferretti A. Effect of manual approaches with osteopathic modality on brain correlates of interoception: an fMRI study. Sci Rep. 2020;10(1):3214.

    PubMed  PubMed Central  Google Scholar 

  278. Yao C, Ren J, Huang R, Tang C, Cheng Y, Lv Z, Kong L, Fang S, Tao J, Fu Y, et al. Transcriptome profiling of microRNAs reveals potential mechanisms of manual therapy alleviating neuropathic pain through microRNA-547-3p-mediated Map4k4/NF-κb signaling pathway. J Neuroinflammation. 2022;19(1):211.

    CAS  PubMed  PubMed Central  Google Scholar 

  279. Schulze NB, Salemi MM, de Alencar GG, Moreira MC, de Siqueira GR. Efficacy of manual therapy on pain, impact of disease, and quality of life in the treatment of fibromyalgia: a systematic review. Pain Physician. 2020;23(5):461–76.

    PubMed  Google Scholar 

  280. Falsiroli Maistrello L, Rafanelli M, Turolla A. Manual therapy and quality of life in people with headache: systematic review and meta-analysis of randomized controlled trials. Curr Pain Headache Rep. 2019;23(10):78.

    PubMed  Google Scholar 

  281. Maher C, McLachlan A, Shaheed CA. A second look at the risks of serious adverse events with orthopaedic manual therapy, paracetamol, and NSAID treatment of neck pain. J Orthop Sports Phys Ther. 2023;0(6):1–2.

    PubMed  Google Scholar 

  282. Kuligowski T, Skrzek A, Cieślik B. Manual therapy in cervical and lumbar radiculopathy: a systematic review of the literature. Int J Environ Res Public Health. 2021;18(11):6176.

    PubMed  PubMed Central  Google Scholar 

  283. Hertzman-Miller RP, Morgenstern H, Hurwitz EL, Yu F, Adams AH, Harber P, Kominski GF. Comparing the satisfaction of low back pain patients randomized to receive medical or chiropractic care: results from the UCLA low-back pain study. Am J Public Health. 2002;92(10):1628–33.

    PubMed  PubMed Central  Google Scholar 

  284. Corcoran KL, Bastian LA, Gunderson CG, Steffens C, Brackett A, Lisi AJ. Association between chiropractic use and opioid receipt among patients with spinal pain: a systematic review and meta-analysis. Pain Med. 2020;21(2):e139–45.

    PubMed  Google Scholar 

  285. Maltese PE, Michelini S, Baronio M, Bertelli M. Molecular foundations of chiropractic therapy. Acta Biomed. 2019;90(10-s):93–102.

    CAS  PubMed  PubMed Central  Google Scholar 

  286. Gevers-Montoro C, Romero-Santiago M, Losapio L, Conesa-Buendía FM, Newell D, Álvarez-Galovich L, Piché M, Ortega-De MA. Presence of tumor necrosis factor-alpha in urine samples of patients with chronic low back pain undergoing chiropractic care: preliminary findings from a prospective cohort study. Front Integr Neurosci. 2022;16:879083.

    CAS  PubMed  PubMed Central  Google Scholar 

  287. Lystad RP, Pollard H. Functional neuroimaging: a brief overview and feasibility for use in chiropractic research. J Can Chiropr Assoc. 2009;53(1):59–72.

    PubMed  PubMed Central  Google Scholar 

  288. Ernst E. Chiropractic treatment for fibromyalgia: a systematic review. Clin Rheumatol. 2009;28(10):1175–8.

    PubMed  Google Scholar 

  289. Bryans R, Descarreaux M, Duranleau M, Marcoux H, Potter B, Ruegg R, Shaw L, Watkin R, White E. Evidence-based guidelines for the chiropractic treatment of adults with headache. J Manipulative Physiol Ther. 2011;34(5):274–89.

    PubMed  Google Scholar 

  290. Yawn BP, Wollan PC, Weingarten TN, Watson JC, Hooten WM, Melton LJ 3rd. The prevalence of neuropathic pain: clinical evaluation compared with screening tools in a community population. Pain Med. 2009;10(3):586–93.

    PubMed  Google Scholar 

  291. Alcantara J, Plaugher G, Klemp DD, Salem C. Chiropractic care of a patient with temporomandibular disorder and atlas subluxation. J Manipulative Physiol Ther. 2002;25(1):63–70.

    PubMed  Google Scholar 

  292. Rubinstein SM. Adverse events following chiropractic care for subjects with neck or low-back pain: do the benefits outweigh the risks? J Manipulative Physiol Ther. 2008;31(6):461–4.

    PubMed  Google Scholar 

  293. Weis CA, Pohlman K, Draper C, daSilva-Oolup S, Stuber K, Hawk C. Chiropractic care for adults with pregnancy-related low back, pelvic girdle pain, or combination pain: a systematic review. J Manipulative Physiol Ther. 2020;43(7):714–31.

    PubMed  Google Scholar 

  294. Gudavalli MR, Vining RD, Salsbury SA, Goertz CM. Training and certification of doctors of chiropractic in delivering manual cervical traction forces: results of a longitudinal observational study. J Chiropr Educ. 2014;28(2):130–8.

    PubMed  PubMed Central  Google Scholar 

  295. Gouveia LO, Castanho P, Ferreira JJ. Safety of chiropractic interventions: a systematic review. Spine (Phila Pa 1976). 2009;34(11):E405-13.

    PubMed  Google Scholar 

  296. Frank DL, Khorshid L, Kiffer JF, Moravec CS, McKee MG. Biofeedback in medicine: who, when, why and how? Ment Health Fam Med. 2010;7(2):85–91.

    PubMed  PubMed Central  Google Scholar 

  297. Hallman DM, Olsson EM, von Schéele B, Melin L, Lyskov E. Effects of heart rate variability biofeedback in subjects with stress-related chronic neck pain: a pilot study. Appl Psychophysiol Biofeedback. 2011;36(2):71–80.

    PubMed  Google Scholar 

  298. McKee MG. Biofeedback: an overview in the context of heart-brain medicine. Cleve Clin J Med. 2008;75(Suppl 2):S31–4.

    PubMed  Google Scholar 

  299. Giggins OM, Persson UM, Caulfield B. Biofeedback in rehabilitation. J Neuroeng Rehabil. 2013;10:60.

    PubMed  PubMed Central  Google Scholar 

  300. Angoules AG, Balakatounis KC, Panagiotopoulou KA, Mavrogenis AF, Mitsiokapa EA, Papagelopoulos PJ. Effectiveness of electromyographic biofeedback in the treatment of musculoskeletal pain. Orthopedics. 2008;31(10):e980–4.

  301. Abd-Elwahab Radi I, Tahoon M. More evidence is needed to determine the effectiveness of full occlusion biofeedback splints used for sleep bruxism (SB) and temporomandibular disorder (TMD) pain. J Evid Based Dent Pract. 2021;21(4):101650.

    PubMed  Google Scholar 

  302. Goldenberg JZ, Brignall M, Hamilton M, Beardsley J, Batson RD, Hawrelak J, Lichtenstein B, Johnston BC. Biofeedback for treatment of irritable bowel syndrome. Cochrane Database Syst Rev. 2019;2019(11):CD012530.

    PubMed  PubMed Central  Google Scholar 

  303. Leong KH, Yip HT, Kuo CF, Tsai SY. Treatments of chronic fatigue syndrome and its debilitating comorbidities: a 12-year population-based study. J Transl Med. 2022;20(1):268.

    CAS  PubMed  PubMed Central  Google Scholar 

  304. Wagner B, Steiner M, Huber DFX, Crevenna R. The effect of biofeedback interventions on pain, overall symptoms, quality of life and physiological parameters in patients with pelvic pain: a systematic review. Wien Klin Wochenschr. 2022;134(Suppl 1):11–48.

    PubMed  Google Scholar 

  305. Pizzoli SFM, Marzorati C, Gatti D, Monzani D, Mazzocco K, Pravettoni G. A meta-analysis on heart rate variability biofeedback and depressive symptoms. Sci Rep. 2021;11(1):6650.

    CAS  PubMed  PubMed Central  Google Scholar 

  306. Swanson KS, Gevirtz RN, Brown M, Spira J, Guarneri E, Stoletniy L. The effect of biofeedback on function in patients with heart failure. Appl Psychophysiol Biofeedback. 2009;34(2):71–91.

    PubMed  Google Scholar 

  307. Umek A, Kos A. The role of high performance computing and communication for real-time biofeedback in sport. Math Probl Eng. 2016;2016:4829452.

    Google Scholar 

  308. Mahrer NE, Gold JI. The use of virtual reality for pain control: a review. Curr Pain Headache Rep. 2009;13(2):100–9.

    PubMed  Google Scholar 

  309. Kim H, Gabbard JL. Assessing distraction potential of augmented reality head-up displays for vehicle drivers. Hum Factors. 2022;64(5):852–65.

    PubMed  Google Scholar 

  310. Kokkinara E, Slater M. Measuring the effects through time of the influence of visuomotor and visuotactile synchronous stimulation on a virtual body ownership illusion. Perception. 2014;43(1):43–58.

    PubMed  Google Scholar 

  311. Mehesz E, Karoui H, Strutton PH, Hughes SW. Exposure to an immersive virtual reality environment can modulate perceptual correlates of endogenous analgesia and central sensitization in healthy volunteers. J Pain. 2021;22(6):707–14.

    PubMed  Google Scholar 

  312. Ahmed H, Mushahid H, Shuja MH. Virtual reality therapy: a promising solution to chronic pain management amidst an opioid crisis. J Glob Health. 2023;13:03033.

    PubMed  PubMed Central  Google Scholar 

  313. Phelan I, Furness PJ, Matsangidou M, Babiker NT, Fehily O, Thompson A, Carrion-Plaza A, Lindley SA. Designing effective virtual reality environments for pain management in burn-injured patients. Virtual Real. 2023;27(1):201–15.

    PubMed  Google Scholar 

  314. Gold JI, Belmont KA, Thomas DA. The neurobiology of virtual reality pain attenuation. Cyberpsychol Behav. 2007;10(4):536–44.

    PubMed  Google Scholar 

  315. Bermo MS, Patterson D, Sharar SR, Hoffman H, Lewis DH. Virtual reality to relieve pain in burn patients undergoing imaging and treatment. Top Magn Reson Imaging. 2020;29(4):203–8.

    PubMed  Google Scholar 

  316. Chen B, Moreland J, Zhang J. Human brain functional MRI and DTI visualization with virtual reality. Quant Imaging Med Surg. 2011;1(1):11–6.

    PubMed  PubMed Central  Google Scholar 

  317. Botella C, Garcia-Palacios A, Vizcaíno Y, Herrero R, Baños RM, Belmonte MA. Virtual reality in the treatment of fibromyalgia: a pilot study. Cyberpsychol Behav Soc Netw. 2013;16(3):215–23.

    PubMed  Google Scholar 

  318. Shiri S, Feintuch U, Weiss N, Pustilnik A, Geffen T, Kay B, Meiner Z, Berger I. A virtual reality system combined with biofeedback for treating pediatric chronic headache–a pilot study. Pain Med. 2013;14(5):621–7.

    PubMed  Google Scholar 

  319. Groninger H, Stewart D, Wesley D, Cowgill J, Mete M. Virtual reality for management of cancer pain: Study rationale and design. Contemp Clin Trials Commun. 2022;26:100895.

    PubMed  PubMed Central  Google Scholar 

  320. Ortiz-Catalan M, Guðmundsdóttir RA, Kristoffersen MB, Zepeda-Echavarria A, Caine-Winterberger K, Kulbacka-Ortiz K, Widehammar C, Eriksson K, Stockselius A, Ragnö C, et al. Phantom motor execution facilitated by machine learning and augmented reality as treatment for phantom limb pain: a single group, clinical trial in patients with chronic intractable phantom limb pain. Lancet. 2016;388(10062):2885–94.

    PubMed  Google Scholar 

  321. Brea-Gómez B, Torres-Sánchez I, Ortiz-Rubio A, Calvache-Mateo A, Cabrera-Martos I, López-López L, Valenza MC. Virtual reality in the treatment of adults with chronic low back pain: a systematic review and meta-analysis of randomized clinical trials. Int J Environ Res Public Health. 2021;18(22):11806.

    PubMed  PubMed Central  Google Scholar 

  322. Wu J, Sun Y, Zhang G, Zhou Z, Ren Z. Virtual reality-assisted cognitive behavioral therapy for anxiety disorders: a systematic review and meta-analysis. Front Psychiatry. 2021;12:575094.

    PubMed  PubMed Central  Google Scholar 

  323. Regan EC, Price KR. The frequency of occurrence and severity of side-effects of immersion virtual reality. Aviat Space Environ Med. 1994;65(6):527–30.

    CAS  PubMed  Google Scholar 

  324. Martini M. Real, rubber or virtual: The vision of “one’s own” body as a means for pain modulation. A narrative review. Conscious Cogn. 2016;43:143–51.

    PubMed  Google Scholar 

  325. O’Connell NE, Marston L, Spencer S, DeSouza LH, Wand BM. Non-invasive brain stimulation techniques for chronic pain. Cochrane Database Syst Rev. 2018;3(3):Cd008208.

    PubMed  Google Scholar 

  326. Mori F, Ribolsi M, Kusayanagi H, Monteleone F, Mantovani V, Buttari F, Marasco E, Bernardi G, Maccarrone M, Centonze D. TRPV1 channels regulate cortical excitability in humans. J Neurosci. 2012;32(3):873–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  327. Ni Z, Müller-Dahlhaus F, Chen R, Ziemann U. Triple-pulse TMS to study interactions between neural circuits in human cortex. Brain Stimul. 2011;4(4):281–93.

    PubMed  Google Scholar 

  328. Dos Santos MF, Oliveira AT, Ferreira NR, Carvalho ACP, Rosado de Castro PH. The contribution of endogenous modulatory systems to TMS- and tDCS-induced analgesia: evidence from PET studies. Pain Res Manag. 2018;2018:2368386.

    Google Scholar 

  329. Li Y, Li W, Zhang T, Zhang J, Jin Z, Li L. Probing the role of the right inferior frontal gyrus during pain-related empathy processing: evidence from fMRI and TMS. Hum Brain Mapp. 2021;42(5):1518–31.

    PubMed  Google Scholar 

  330. Lin C, Lee SH, Weng HH. Gray matter atrophy within the default mode network of fibromyalgia: a meta-analysis of voxel-based morphometry studies. Biomed Res Int. 2016;2016:7296125.

    PubMed  PubMed Central  Google Scholar 

  331. Feng X, Niu L, Long M, Luo K, Huang X, Chen M, Lin Z, Zhou W, Yi S, Ao L. Transcranial ultrasound stimulation of the anterior cingulate cortex reduces neuropathic pain in mice. Evid Based Complement Alternat Med. 2021;2021:6510383.

    PubMed  PubMed Central  Google Scholar 

  332. Busch V, Zeman F, Heckel A, Menne F, Ellrich J, Eichhammer P. The effect of transcutaneous vagus nerve stimulation on pain perception–an experimental study. Brain Stimul. 2013;6(2):202–9.

    PubMed  Google Scholar 

  333. Guo W, He Y, Zhang W, Sun Y, Wang J, Liu S, Ming D. A novel non-invasive brain stimulation technique: “Temporally interfering electrical stimulation.” Front Neurosci. 2023;17:1092539.

    PubMed  PubMed Central  Google Scholar 

  334. Jiang X, Yan W, Wan R, Lin Y, Zhu X, Song G, Zheng K, Wang Y, Wang X. Effects of repetitive transcranial magnetic stimulation on neuropathic pain: a systematic review and meta-analysis. Neurosci Biobehav Rev. 2022;132:130–41.

    PubMed  Google Scholar 

  335. Stilling JM, Monchi O, Amoozegar F, Debert CT. Transcranial Magnetic and Direct Current Stimulation (TMS/tDCS) for the treatment of headache: a systematic review. Headache. 2019;59(3):339–57.

    PubMed  Google Scholar 

  336. Emami Zeydi A, Esmaeili R, Hasanzadeh Kiabi F, Sharifi H. Repetitive transcranial magnetic stimulation as a promising potential therapeutic modality for the management of cancer-related pain: an issue that merits further research. Indian J Palliat Care. 2017;23(1):109–10.

    PubMed  PubMed Central  Google Scholar 

  337. Lefaucheur JP. Use of repetitive transcranial magnetic stimulation in pain relief. Expert Rev Neurother. 2008;8(5):799–808.

    PubMed  Google Scholar 

  338. Teixeira PEP, Alawdah L, Alhassan HAA, Guidetti M, Priori A, Papatheodorou S, Fregni F. The analgesic effect of Transcranial Direct Current Stimulation (tDCS) combined with physical therapy on common musculoskeletal conditions: a systematic review and meta-analysis. Princ Pract Clin Res. 2020;6(1):23–6.

    PubMed  PubMed Central  Google Scholar 

  339. Mendonca ME, Simis M, Grecco LC, Battistella LR, Baptista AF, Fregni F. Transcranial direct current stimulation combined with aerobic exercise to optimize analgesic responses in fibromyalgia: a randomized placebo-controlled clinical trial. Front Hum Neurosci. 2016;10:68.

    PubMed  PubMed Central  Google Scholar 

  340. de Berker AO, Bikson M, Bestmann S. Predicting the behavioral impact of transcranial direct current stimulation: issues and limitations. Front Hum Neurosci. 2013;7:613.

    PubMed  PubMed Central  Google Scholar 

  341. Pikhovych A, Stolberg NP, Jessica Flitsch L, Walter HL, Graf R, Fink GR, Schroeter M, Rueger MA. Transcranial direct current stimulation modulates neurogenesis and microglia activation in the mouse brain. Stem Cells Int. 2016;2016:2715196.

    PubMed  PubMed Central  Google Scholar 

  342. Dedoncker J, Brunoni AR, Baeken C, Vanderhasselt MA. A systematic review and meta-analysis of the effects of Transcranial Direct Current Stimulation (tDCS) over the dorsolateral prefrontal cortex in healthy and neuropsychiatric samples: influence of stimulation parameters. Brain Stimul. 2016;9(4):501–17.

    PubMed  Google Scholar 

  343. Langevin HM, Schnyer R, MacPherson H, Davis R, Harris RE, Napadow V, Wayne PM, Milley RJ, Lao L, Stener-Victorin E, et al. Manual and electrical needle stimulation in acupuncture research: pitfalls and challenges of heterogeneity. Altern Complement Med. 2015;21(3):113–28.

    Google Scholar 

  344. Good M, Picot BL, Salem SG, Chin C-C, Picot SF, Lane D. Cultural differences in music chosen for pain relief. J Holist Nurs. 2000;18(3):245–60.

    CAS  PubMed  Google Scholar 

  345. Meier M, Unternaehrer E, Dimitroff SJ, Benz ABE, Bentele UU, Schorpp SM, Wenzel M, Pruessner JC. Standardized massage interventions as protocols for the induction of psychophysiological relaxation in the laboratory: a block randomized, controlled trial. Sci Rep. 2020;10(1):14774.

    CAS  PubMed  PubMed Central  Google Scholar 

  346. Turi Z, Normann C, Domschke K, Vlachos A. Transcranial magnetic stimulation in psychiatry: is there a need for electric field standardization? Front Hum Neurosci. 2021;15:639640.

    PubMed  PubMed Central  Google Scholar 

  347. Mathur AS. Low cost virtual reality for medical training. In: 2015 IEEE Virtual Reality (VR); 2015.

  348. Olshannikova E, Ometov A, Koucheryavy Y, Olsson T. Visualizing big data with augmented and virtual reality: challenges and research agenda. J Big Data. 2015;2(1):22.

    Google Scholar 

  349. Cocchi L, Zalesky A. Personalized transcranial magnetic stimulation in psychiatry. Biol Psychiatry Cogn Neurosci Neuroimaging. 2018;3(9):731–41.

    PubMed  Google Scholar 

  350. Kosmyna N, Maes P. AttentivU: an eeg-based closed-loop biofeedback system for real-time monitoring and improvement of engagement for personalized learning. Sensors. 2019;19(23):5200.

    PubMed  PubMed Central  Google Scholar 

  351. Liu J, Quan S, Zhao L, Yuan K, Wang Y, Zhang Y, Wang Z, Sun M, Hu L. Evaluation of a clustering approach to define distinct subgroups of patients with migraine to select electroacupuncture treatments. Neurology. 2023;101(7):e699–709.

    PubMed  PubMed Central  Google Scholar 

  352. Guo H, Que M, Shen J, Nie Q, Chen Y, Huang Q, Jin A. Effect of music therapy combined with free position delivery on labor pain and birth outcomes. Appl Bionics Biomech. 2022;2022:8963656.

    PubMed  PubMed Central  Google Scholar 

  353. You WD, Fu DG, Yu HY. A study of pulsed radiofrequency combined with never block in the treatment of postherpeticn euralgia (Chin). Pract J Cardiocereb Pneumovasc Dis. 2011;19(3):477–9.

    Google Scholar 

  354. Zhang Q, Hu S, Talay R, Xiao Z, Rosenberg D, Liu Y, Sun G, Li A, Caravan B, Singh A, et al. A prototype closed-loop brain-machine interface for the study and treatment of pain. Nat Biomed Eng. 2023;7(4):533–45.

    PubMed  Google Scholar 

  355. Mekhail N, Levy RM, Deer TR, Kapural L, Li S, Amirdelfan K, Hunter CW, Rosen SM, Costandi SJ, Falowski SM, et al. Long-term safety and efficacy of closed-loop spinal cord stimulation to treat chronic back and leg pain (Evoke): a double-blind, randomised, controlled trial. Lancet Neurol. 2020;19(2):123–34.

    PubMed  Google Scholar 

  356. Moore RA, Derry S, Wiffen PJ. Challenges in design and interpretation of chronic pain trials. Br J Anaesth. 2013;111(1):38–45.

    CAS  PubMed  Google Scholar 

  357. Gewandter JS, Dworkin RH, Turk DC, Farrar JT, Fillingim RB, Gilron I, Markman JD, Oaklander AL, Polydefkis MJ, Raja SN, et al. Research design considerations for chronic pain prevention clinical trials: IMMPACT recommendations. Pain Rep. 2021;6(1):e895.

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We gratefully acknowledge all researchers in this field whose work provided the foundation for the evidence reviewed in this manuscript.

Funding

This work was supported by National Natural Science Foundation of China (NNSFC), China; Contract grant number: 82102645, 82172526, 82372553; Guangdong Basic and Applied Basic Research Foundation, China; Contract grant number: 2021A1515011042, 2019A1515110739; China Postdoctoral Science Foundation, China; Contract grant number: 2019M662995.

Author information

Authors and Affiliations

Authors

Contributions

YS and WW contributed to the conception and design of the study. YS wrote the first draft of the manuscript. YS and WW wrote sections of the manuscript. All authors contributed to manuscript revision, read, and approved the submitted version. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Wen Wu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Supplementary Information

Additional file 1: Figure S1.

Clinical applications of physical modalities for chronic pain conditions. Note: Chronic pain conditions in the figure refer to ICD-11 classifications. Examples given in the article include specific diseases falling into these categories. Figure S2. Clinical applications of psychological interventions for chronic pain conditions. Note: Chronic pain conditions in the figure refer to ICD-11 classifications. Examples given in the article include specific diseases falling into these categories. Figure S3. Clinical applications of complementary and alternative therapies for chronic pain conditions. Note: Chronic pain conditions in the figure refer to ICD-11 classifications. Examples given in the article include specific diseases falling into these categories. Figure S4. Clinical applications of others therapies for chronic pain conditions. Note: Chronic pain conditions in the figure refer to ICD-11 classifications. Examples given in the article include specific diseases falling into these categories.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shi, Y., Wu, W. Multimodal non-invasive non-pharmacological therapies for chronic pain: mechanisms and progress. BMC Med 21, 372 (2023). https://doi.org/10.1186/s12916-023-03076-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12916-023-03076-2

Keywords