Contents
- Spinal cord stimulator
- Spinal Cord Stimulator Surgery
- Spinal cord stimulator complications
- What is Chronic Pain?
- What is Complex Regional Pain Syndrome?
- Complex regional pain syndrome causes
- Risk factors for developing complex regional pain syndrome
- Complex regional pain syndrome prevention
- Complex regional pain syndrome symptoms
- Complex regional pain syndrome complications
- Complex regional pain syndrome diagnosis
- Complex regional pain syndrome treatment
- Rehabilitation and physical therapy and occupational therapy
- Psychotherapy
- Complex regional pain syndrome medications
- Sympathetic nerve block
- Surgical sympathectomy
- Spinal cord stimulation
- Dorsal root ganglion stimulation
- Other types of neural stimulation
- Epidural infusion
- Intrathecal drug pumps
- Topical Treatments
- Emerging treatments
- Amputation
- Living with complex regional pain syndrome
Spinal cord stimulator
Spinal cord stimulator is an implanted spinal cord stimulation device that sends low levels of electricity directly into your spinal cord to relieve pain. Spinal cord stimulator consists of thin wires (the electrodes) and a small, pacemaker-like battery pack (the generator). The electrodes are placed between your spinal cord and the vertebrae (the epidural space), and the generator is placed under your skin, usually near your buttocks or abdomen. Spinal cord stimulation is used most often after nonsurgical pain treatment options have failed to provide sufficient relief. Spinal cord stimulators require two procedures to test and implant the device: the trial and the implantation.
Spinal cord stimulator is a long-lasting procedure to treat nerve pain problems. A device is implanted into the spine, and a remote control is used to send low levels of electricity. It causes tingling against the nerves in the back or neck, which reduces the amount of pain felt.
The procedure is done in two steps, with the first step as a trial to see if it provides relief. If pain relief is above 60%, the second step is to permanently implant the device.
Experts still don’t fully understand the mechanisms behind spinal cord stimulation, but they know that spinal cord stimulator may target multiple muscle groups directly from the spine and even alter how the brain senses pain.
Traditional spinal cord stimulators replace the sensation of pain with light tingling, called paresthesia. For patients who find these paresthesiae uncomfortable, newer devices offer “sub-perception” stimulation that cannot be felt.
Many of the latest devices are placed by physicians with highly specialized training in interventional pain management under X-ray and/or ultrasound guidance.
Spinal cord stimulation results in inadequate pain relief in about 50% of patients 1. Tolerance can also occur in up to 20 to 40 percent of patients. The effectiveness of the spinal cord stimulation decreases over time 2.
Spinal cord stimulator types
Spinal cord stimulators come in 3 main types:
- Conventional implantable pulse generator (IPG) is a battery-operated spinal code stimulator. A battery is placed in the spine during an operation. When it runs out, the battery must be replaced with another surgery. This device can be a good choice for people with pain in just one body part because it has a lower electrical output.
- Rechargeable implantable pulse generator (IPG) works similarly to the conventional spinal code stimulator, with the difference that the battery can be recharged without another surgery. Because the energy source is rechargeable, these spinal code stimulators can put out more electricity. This may be a better choice for people with pain in the lower back or in one or both legs, as the electrical signal can reach further.
- Radiofrequency stimulator uses a battery that’s outside the body. Radiofrequency stimulator is rarely used today because of newer designs and better technology. It has rechargeable batteries, and like the rechargeable implantable pulse generator (IPG), it may be better for people with pain in the lower back and legs because of the device’s power.
Your surgeon will explain how to operate the device and adjust the intensity of the electrical signal, which all three types of stimulators support. Different body positions may require different stimulator settings, such as one setting that works better for sitting and another for walking. To help you easily access the most used settings, most devices allow doctors to save two or three preset programs. Some newer devices feature several waveforms for electricity delivery, including high frequency, burst and high-density stimulation.
What is spinal cord stimulation used for?
Spinal cord stimulation is used most often after nonsurgical pain treatment options have failed to provide sufficient relief. Spinal cord stimulators may be used to treat or manage different types of chronic pain, including:
- Back pain, especially back pain that continues even after surgery (failed back surgery syndrome)
- Post-surgical pain
- Arachnoiditis (painful inflammation of the arachnoid, a thin membrane that covers the brain and spinal cord)
- Heart pain (angina) untreatable by other means
- Injuries to the spinal cord
- Nerve-related pain (such as severe diabetic neuropathy and cancer-related neuropathy from radiation, surgery or chemotherapy)
- Peripheral vascular disease
- Complex regional pain syndrome
- Pain after an amputation
- Visceral abdominal pain and perineal pain
Spinal cord stimulation can improve overall quality of life and sleep, and reduce the need for pain medicines. It is typically used along with other pain management treatments, including medications, exercise, physical therapy and relaxation methods.
Who should get a spinal cord stimulator?
As with all treatments, your doctor will want to make sure spinal cord stimulation is right for you and that it is likely to provide significant relief from your chronic pain. To make this recommendation, your pain specialist will likely order imaging tests and psychological screening. Some insurance companies require psychological screening to ensure disorders like depression or anxiety aren’t worsening your pain.
Each patient is different, but generally, people who benefit the most from spinal cord stimulation are those who:
- Have not experienced sufficient pain relief with medications, less-invasive therapies or prior surgeries
- Do not have psychiatric disorders that would decrease the effectiveness of the procedure
Why Spinal cord stimulation Procedure is Performed?
Your doctor may recommend spinal cord stimulation if you have:
- Back pain that continues or gets worse, even after surgery to correct it
- Complex regional pain syndrome (CRPS)
- Long-term (chronic) back pain, with or without arm or leg pain
- Nerve pain or numbness in the arms or legs
- Swelling (inflammation) of the lining of the brain and spinal cord
Spinal cord stimulator is used after you have tried other treatments such as medicines and exercise and they have not worked.
The main components include an implanted signal generator that is connected to one or two implanted leads, and externally worn transmitter that is doctor and patient controlled. The therapy utilizes pulsed electrical current to create an energy field that acts on nerves near the spinal column to block nerve impulses in the spine 3.
Stimulator placement is done in two stages. A trial (test) stage is done to see how well spinal cord stimulation works for you. If the trial stage is a success, the permanent stimulator system is put into place.
Spinal cord stimulator contraindications
Pacemakers and defibrillators are often compatible with spinal cord stimulator implants 4. However, a history of a previous cardiac pacemaker or cardiac defibrillator requires the approval of spinal cord stimulator implantation by a cardiologist before the procedure 4. Having either a cardiac pacemaker or defibrillator remains a relative contraindication to spinal cord stimulator implant. Both devices require close follow up following spinal cord stimulator implants 5, 6, 7. Severe low platelet count (thrombocytopenia) or uncontrolled bleeding disorder (coagulopathy) are also contraindications to spinal cord stimulator implant due to increased risk of a spinal epidural hematoma 4. Active infection is an absolute contraindication to implantation of a spinal cord stimulator 4. There is limited information regarding spinal cord stimulator implantation in a pregnant patient 8. Although, given the need for fluoroscopy to guide the spinal cord stimulator procedure, the procedure is almost always postponed until birth.
Spinal Cord Stimulator Surgery
Spinal cord stimulators require two procedures to test and implant the device: the trial and the implantation.
Spinal cord stimulator trial
The first step is a trial period. Your surgeon will implant a temporary device for you to test out. Your skin will be numbed with a local anesthetic. Guided by a specific type of X-ray called fluoroscopy, your surgeon will carefully insert the electrodes (wires) in the epidural space of your spine. The location of your pain affects where these electrodes will be placed along the spine. Your surgeon may ask for your feedback during the procedure to best position the electrodes. This trial procedure typically requires only one incision in your lower back to place the electrodes (wires). These electrodes (wires) will be connected to a small current generator/battery outside of your body that you carry like a cell phone typically on a belt, you’ll wear around your waist. The procedure takes about 1 hour. You will be able to go home after the leads are placed.
If the treatment greatly reduces your pain, you will be offered a permanent generator. The generator will be implanted a few weeks later.
- You will be asleep and pain-free with general anesthesia.
- The generator will be inserted under the skin of your abdomen or buttocks through a small surgical cut.
- The procedure takes about 1 to 2 hours.
The generator runs on batteries. Some batteries are rechargeable. Others last 2 to 5 years. You will need another surgery to replace the battery.
For about a week, you will evaluate how well the device reduces your pain. The trial is considered a success if you experience a 50% or greater reduction in pain level.
If unsuccessful, the wires can easily be removed in the clinic without damage to the spinal cord or nerves. If successful, surgery is scheduled to permanently implant the device.
Spinal Cord Stimulator Implantation
During the permanent implantation procedure, the generator is placed underneath the skin and the trial electrodes are replaced with sterile electrodes. Unlike the trial electrodes, these will be anchored by sutures to minimize movement.
The implantation can take about 1-2 hours and is typically performed as an outpatient procedure.
After the local anesthesia has been administered, your surgeon will make one incision (typically along your lower abdomen or buttocks) to hold the generator and another incision (along your spine) to insert the permanent electrodes. The incisions are about the length of a driver’s license. As in the trial procedure, fluoroscopy is used to determine where the electrodes are placed.
Once the electrodes and generator are connected and running, your surgeon will close the incisions.
Your surgeon may provide sedation to keep you comfortable and ask for your feedback during placement of the electrodes.
After the Procedure
After the permanent generator is placed, the surgical cut will be closed and covered with a dressing. You will be taken to the recovery room to wake up from the anesthesia.
Most people can go home the same day, but your surgeon may want you to stay overnight in the hospital. You will be taught how to care for your surgical site.
You should avoid heavy lifting, bending, and twisting while you are healing. Light exercise such as walking can be helpful during recovery.
- After the procedure you may have less back pain and will not need to take as much pain medicines. But, the treatment does not cure back pain or treat the source of the pain.
Spinal cord stimulator complications
Risks of this surgery include any of the following 3:
- Cerebrospinal fluid (CSF) leakage
- Damage to the nerves that come out of the spine, causing paralysis, weakness, or pain that does not go away
- Infection of the battery or electrode site (if this occurs, the hardware usually needs to be removed)
- Movement of or damage to the generator or leads that requires more surgery
- Pain after surgery
- Problems with how the stimulator works, such as sending too strong of a signal, stopping and starting, or sending a weak signal
- The stimulator may not work
The spinal cord stimulator device may interfere with other devices, such as pacemakers and defibrillators. After the spinal cord stimulator is implanted, you may not be able to get an MRI anymore. Discuss this with your doctor.
Complication rates for spinal cord stimulators are high, ranging from 5% up to 40% 9, 10. Most commonly, lead migration occurs, causing inadequate pain relief and requiring revision surgery and anchoring 11, 12. Lead movement often occurs in the cervical region of the spinal cord, given an increased range of motion of the cervical vertebra 13, 14. Spinal cord stimulator lead fracture can occur in up to 9% of placements 15, 16. Seromas are also very common and may require surgical incision and drainage 9, 17. The risk of infection following a spinal cord stimulator placement is between 2.5% and 12% 18, 19. Direct spinal cord trauma could occur. The most significant infectious complication would be a spinal cord abscess. Dural puncture is rare but can cause a post-dural headache in up to 70% of patients 17, 20, 21. The most critical adverse event in spinal cord stimulator placement would be a spinal epidural hematoma, which requires immediate neurosurgical decompression. The incidence of a spinal epidural hematoma is 0.71% 22.
What is Chronic Pain?
Chronic pain also called chronic pain syndrome (CPS) or persistent pain is an umbrella term referring to pain that lasts 3 to 6 months or pain that persists past “normal healing time” 23, 24, 25, 26, 27, 28, 29, 30, 23, 31. The International Association for the Study of Pain (IASP) defines “chronic pain” as persistent or recurrent pain lasting longer than 3 months 23, 24. Unlike acute pain, which is temporary, chronic pain can be constant or come and go, often interfering with daily life and affecting your physical and emotional well-being 32. It can stem from an ongoing health condition or persist even after the initial cause has been treated or is no longer present.
Pain is an unpleasant physical feeling, such as a prick, tingle, sting, burn, or ache. Pain may be sharp or dull. You may feel pain in one area of your body, or all over.
There are two types of pain: acute pain and chronic pain 33.
- Acute pain happens suddenly, starts out sharp or intense, and serves as a warning sign of disease or threat to the body. Acute pain lets you know that you may be injured or a have problem you need to take care of. Acute pain is caused by injury, surgery, illness, trauma, or painful medical procedures and generally lasts from a few minutes to less than six months. Acute pain usually disappears whenever the underlying cause is treated or healed.
- Chronic pain is different. Chronic pain usually doesn’t have a useful purpose. The pain may last for weeks, months, or even years. The original cause may have been an injury or infection. There may be an ongoing cause of pain, such as arthritis or cancer. In some cases there is no clear cause. Environmental and psychological factors can make chronic pain worse. Chronic pain can significantly affect a person’s ability to perform daily activities, work, mood and engage in social interactions. Chronic pain can also affect your sleep leading to exhaustion and psychological stress.
- The transition from acute to chronic pain may be understood as a series of relatively discrete changes in your brain and spinal cord (central nervous system) 34, 35, 36, 37, 38, 28. Chronic pain involves the activation of secondary mechanisms such as the sensitization of second-order neurons by upregulation of N-methyl-D-aspartic acid (NMDA) channels and alteration in microglia cytoarchitecture. Chronic pain, with its multiple factors for perpetuation, often benefits from a multidisciplinary approach to treatment 39.
- Acute pain and chronic pain need to be treated differently. Acute pain can often be relieved with painkillers such as acetylsalicylic acid (Aspirin), ibuprofen and paracetamol (acetaminophen). But these drugs don’t work in many people who have chronic pain. Even strong painkillers such as opioids often don’t help chronic pain.
Pain is also personal and subjective, and individuals may perceive and describe it differently, meaning what’s painful for one person may not be painful for another person. The experience of chronic pain is highly individual, with different people experiencing varying levels of intensity and impact. This variation makes personalized treatment plans essential for effective management.
Pain research has led to some discoveries that might be surprising 33:
- The fear of pain can be painful. Even if your body isn’t in any real danger and your tissue and organs are healthy, you may still feel pain – if your brain thinks you’re in danger. The purpose of this kind of pain is to help you avoid the thing that is thought to be dangerous. One aim of modern pain management is to reduce the fear of pain – for example, the fear of pain caused by exercising too much or doing the wrong kind of exercise.
- Pain that has no physical cause is still “real”. People who have pain for no known physical reason are sometimes accused of imagining it. That’s not true, though. Their pain just arises in a different way. A scientific experiment made this clearer: healthy participants agreed to wear a cap that sends out electrical impulses that cause headaches or so they were told. In actual fact, the cap didn’t send out any impulses or have any other effects. But the people in the experiment still felt pain and the higher the “pain impulse” dial was turned up, the worse their pain became. The participants didn’t imagine the pain, though. They really felt it. This is because their brains thought they were being exposed to danger, so it produced pain signals.
- The severity of pain often isn’t directly related to the extent of physical damage. Very severe injuries like gunshot wounds don’t always cause immediate pain. Severe pain would make it harder for the wounded person to get out of danger in order to survive. Another example, some people have severe damage to their spine caused by wear and tear but it doesn’t hurt and some people have really bad back pain although no damage can be seen on x-ray images.
- Pain depends on outside factors. Pain severity can be influenced by how dangerous your brain considers something to be. This can be shown by the following experiment: A group of people volunteered to have a very cold metal pin (minus 20 degrees Celsius, which is about minus 4 degrees Fahrenheit) held against their hand for half a second. The metal pin either had a red light or a blue light on it. People who saw a red light felt more severe pain than people who saw a blue light – even though the temperature of the metal pin was the same, and it was held against their hand for the same amount of time. This is because we tend to associate the color red with danger and extreme heat, and see it as a more threatening color.
- Pain is influenced by how you feel. For instance, people feel less pain if their doctor takes more time for them and explains what is causing the pain. And pain may feel worse on a stressful day at work than at a weekend when out and about with good friends. Last, but not least, positive thoughts, pleasant experiences or music can distract you from pain or even allow you to forget it for a while.
These things are important for people who have chronic pain. They show us that your experience of pain can be influenced by what your brain associates with it. For instance, fear of pain plays an important role. Understanding what causes chronic pain may also help people to cope with it better.
Common chronic pain complaints include headache, low back pain, cancer pain, arthritis pain, neurogenic pain (pain resulting from damage to the peripheral nerves or to the central nervous system itself), psychogenic pain (pain not due to past disease or injury or any visible sign of damage inside or outside the nervous system). A person may have two or more co-existing chronic pain conditions. Such conditions can include chronic fatigue syndrome, endometriosis, fibromyalgia, inflammatory bowel disease, interstitial cystitis, temporomandibular joint dysfunction, and vulvodynia. It is not known whether these disorders share a common cause.
Depression and stress tend to make pain worse, including chronic pain.
Many older adults have chronic pain. Women also report having more chronic pain than men, and they are at a greater risk for many pain conditions. Some people have two or more chronic pain conditions.
Chronic pain can occur anywhere in the body. People with chronic pain complain of 40:
- headaches
- back pain
- cancer pain
- arthritis pain
- pain resulting from nerve damage.
The pain can be described as 40:
- a dull ache
- soreness
- stiffness
- stinging
- squeezing
- throbbing
- burning
- shooting.
Sometimes people with chronic pain have other symptoms. These could include feeling tired, having trouble sleeping, or mood changes. The pain itself often leads to other symptoms. These include low self-esteem, anger, depression, anxiety, or frustration.
Chronic pain is not always curable, but treatments can help. They include 41:
- Pain Relievers and other medicines
- Acupuncture and massage
- Electrical stimulation e.g. spinal cord stimulator, brain stimulation
- Surgery
- Physical therapy
- Psychotherapy
- Relaxation and meditation therapy
- Biofeedback
- Behavior modification
- Some physicians use placebos, which in some cases has resulted in a lessening or elimination of pain.
Table 1. Chronic Pain Classification (11th version of the International Classification of Diseases (ICD-11))
Pain Type | Definition | Neurobiologic Mechanism | Examples |
---|---|---|---|
Chronic Primary Pain | |||
1. Chronic widespread pain | Widespread pain persisting for longer than 3 months, associated with emotional distress or functional disability | Central sensitization | Fibromyalgia |
2. Complex Regional Pain Syndrome (CRPS) | Disorder of body region, usually distal limbs, characterized by pain (allodynia), swelling, loss of function, vasomotor instability, skin changes | Neuropathic Central sensitization | Chronic Regional Pain Syndrome (formerly reflex sympathetic dystrophy) |
3. Chronic primary headache or orofacial pain | Idiopathic headache or orofacial pain, not secondary to another condition | Nociceptive Neuropathic Central sensitization | Chronic migraine or temporomandibular disorder |
4. Chronic primary visceral pain | Persistent or recurrent pain originating from internal organs, without a clear organic cause | Central sensitization | Irritable bowel syndrome |
5. Chronic primary musculoskeletal pain | Chronic pain experienced in muscles, bones, joints, or tendons that cannot be attributed directly to a known disease or tissue damage process 42 | Nociceptive Neuropathic Central sensitization | Non-specific low back pain |
Chronic Secondary Pain | |||
1. Chronic cancer-related pain | Pain caused by the cancer itself (by the primary tumor or by metastases) or by its treatment (surgery, chemotherapy, or radiotherapy) 43 | Nociceptive Neuropathic Central sensitization | Chronic cancer pain, chronic cancer treatment pain (eg, chemotherapy-induced peripheral neuropathy, radiation fibrosis) |
2. Chronic postsurgical or posttraumatic pain | Pain secondary to surgery or trauma which persists for longer than 3 months | Nociceptive Neuropathic Central sensitization | Incisional pain, nerve injury due to trauma or surgery (eg, persistent whiplash or low back pain after trauma) |
3. Chronic neuropathic pain | Pain caused by a lesion or disease of the somatosensory nervous system 44 | Neuropathic | Trigeminal neuralgia, chronic painful polyneuropathy (eg, diabetic polyneuropathy), postherpetic neuralgia |
4. Chronic secondary headache or orofacial pain | Headaches or orofacial pains, secondary to a medical condition | Nociceptive Neuropathic Central sensitization | Head/face pain secondary to trauma, tumor, hemorrhage, etc. |
5. Chronic secondary visceral pain | Persistent or recurrent pain originating from internal organs, due to a secondary cause | Nociceptive | Abdominal pain due to adhesions or ischemia |
6. Chronic secondary musculoskeletal pain | Persistent or recurrent pain that arises as part of a disease process directly affecting bones, joints, muscles, or related soft tissues | Nociceptive | Rheumatoid arthritis, osteoarthritis |
Chronic pain causes
The cause of chronic pain is multifactorial and complex and still is poorly understood 45, 46, 47, 26. The development and chronicization of pain can be also influenced by several psychological and social factors, such as depression, catastrophizing, avoidance behaviors, somatization, responses from significant others and cultural attitudes 48. In addition, chronic pain is often accompanied by several biological (eg, depression of the immune system), physical (eg, impaired functioning), psychological (eg, depression) and social consequences (eg, job loss), which all contribute in aggravating the patient’s burden 49.
The most common causes of chronic pain include 32:
- Injuries
- Arthritis
- Back problems
- Nerve damage
- Fibromyalgia
- Headaches and migraines
- Post-surgical pain
- Cancer
- Infections
- Stress
- Anxiety and depression
- Poor posture and overuse
Certain factors can contribute to an increased risk of chronic pain (female gender, older age, lower socioeconomic status, geographical and cultural background, and genetics 50, 51. Other factors associated with chronic pain conditions are modifiable, such as smoking status, alcohol intake, nutrition, obesity, comorbidities, employment status and occupational factors, and physical activity level 50, 52.
An observational study found that individuals tend to smoke more cigarettes when experiencing higher pain levels and make fewer attempts to quit smoking during such periods 53. This behavior can be attributed to the acute pain-relieving effect of nicotine, which makes it challenging for them to quit due to the pleasurable sensations it provides 54. Despite its short-term pain relief benefits, tobacco smoking is associated with chronic pain intensity and prevalence in the long run 55, 54, 56, 57. This highlights the importance of smoking cessation strategies to improve the individual’s understanding of the relationship between pain and smoking, potentially increasing their commitment to a smoking cessation program 53.
Smoking status in individuals with chronic pain is also associated with alcohol-drug and opioid dependence 58, 59. When smoking and alcohol consumption are combined, their negative effects are compounded due to the alcohol’s ability to stop the body’s capacity to breakdown the cancer causing compounds found in cigarettes 60. Furthermore, it is worth noting that alcohol initially has an acute pain relieving effect 61, 62, which can elevate the risk of alcohol abuse in individuals with chronic pain 61, 63.
Recent studies have shown that chronic pain patients with high levels of pain catastrophizing are more likely to engage in heavy drinking 64.
Some authors have suggested that chronic pain might be a learned behavioral that begins with a noxious stimulus that causes pain. This pain behavior then is rewarded externally or internally. Therefore, this pain behavior is reinforced, and then it occurs without any noxious stimulus. Internal reinforcers are relief from personal factors associated with many emotions (eg, guilt, fear of work, sex, responsibilities). External reinforcers include such factors as attention from family members and friends, socialization with the physician, medications, compensation, and time off from work.
Patients with several psychological syndromes (eg, major depression, somatization disorder, hypochondriasis, conversion disorder) are prone to developing chronic pain syndrome.
Various neuromuscular, reproductive, gastrointestinal (GI), and urologic disorders may cause or contribute to chronic pain. Sometimes multiple contributing factors may be present in a single patient.
In a study by Alonso-Blanco 65, a connection was found in women between the number of active myofascial trigger points (MTrPs) and the intensity of spontaneous pain, as well as widespread mechanical hypersensitivity. Nociceptive inputs from these MTrPs may be linked to central sensitization.
A literature review by Gupta et al 66 indicated that in chronic pain patients, primary sensorimotor structural and functional changes are more prominent in females than in males. Males and females differed with regard to the nature and degree of insula changes (with males showing greater insula reactivity), as well as in the extent of anterior cingulate structural changes and in reactivity to emotional arousal.
Chronic pain symptoms
Chronic pain is an umbrella term referring to pain that lasts 3 to 6 months or pain that persists past “normal healing time” 23, 24, 25, 26, 27.
The most common symptoms of chronic pain include 32:
- Persistent pain
- Fatigue
- Stiffness
- Sleep disturbances
- Mood changes
- Decreased mobility
- Sensitivity to touch
- Appetite changes
- Cognitive difficulties
- Social withdrawal
Common chronic pain syndrome include headache, low back pain, cancer pain, arthritis pain, neurogenic pain (pain resulting from damage to the peripheral nerves or to the central nervous system itself), psychogenic pain (pain not due to past disease or injury or any visible sign of damage inside or outside the nervous system). A person may have two or more co-existing chronic pain conditions, several of which cause pain, may occur together. Such conditions can include chronic fatigue syndrome, endometriosis, fibromyalgia, irritable bowel syndrome, inflammatory bowel disease, interstitial cystitis (painful bladder syndrome), temporomandibular joint dysfunction, and vulvodynia (chronic vulvar pain).
Chronic pain diagnosis
Diagnosing chronic pain typically involves a comprehensive evaluation to understand the underlying causes and assess the impact on the patient’s life. Your medical doctor will likely ask you about your symptoms, the intensity and location of pain, and the history of your injury, surgery, or illness. Young patients may be asked to rate their pain intensity using the faces of pain scale.
The decision to perform any laboratory or imaging evaluations is based on the need to confirm the diagnosis and to rule out other potentially life-threatening illnesses. Sometimes certain investigations are needed to provide appropriate and safe medical or surgical treatment. The recommended treatment should be based on clinical findings or changes in examination findings.
Extreme care should be taken during diagnostic testing for chronic pain syndrome. Carefully review prior testing to eliminate unnecessary repetition.
Routine complete blood count (CBC), urinalysis, and selected tests for suspected disease are important. Urine or blood toxicology is important for drug detoxification, as well as opioid therapy.
Some common diagnostic methods and tests include:
- medical history review
- physical examination
- pain assessment tools
- imaging studies
- x-rays
- MRI
- CT scans
- bone scans
- ultrasound
- electromyography (EMG) and nerve conduction studies
- blood tests
- nerve blocks and injections
- psychological evaluation
Imaging studies
Imaging studies, including with radiography, magnetic resonance imaging (MRI), and computed tomography (CT) scanning, are important tools in the workup of patients with chronic pain syndrome.
Chronic pain treatment
Management of chronic pain in patients with multiple problems is complex, and may require a comprehensive approach involving medical treatment, reducing/tapering or even stopping non-working medications, lifestyle changes, physical therapy, psychological treatment such as cognitive behavioral therapy (CBT) or mindfulness and sometimes alternative treatments like acupuncture and mind-body techniques 67, 68. Physical therapy techniques include hot or cold applications, positioning, stretching exercises, traction, massage, ultrasonographic therapy, transcutaneous electrical nerve stimulation (TENS), and manipulations. Lifestyle modifications, such as a healthy diet, weight management, and sleep hygiene, are also often recommended. Other treatments include nerve blocks, spinal cord stimulation, and intrathecal morphine pumps. The most effective treatment plans are often personalized, combining several of these approaches based on the individual’s specific condition, type of pain, and overall health. The goal is often to reduce pain and most commonly improve function rather than eliminate the pain entirely.
Treatment of chronic pain must be tailored for each individual patient. The treatment should be aimed at interruption of reinforcement of the pain behavior and modulation of the pain response. The goals of treatment must be realistic and should be focused on restoration of normal function (minimal disability), better quality of life, reduction of use of medication, and prevention of relapse of chronic symptoms.
Psychological interventions, in conjunction with medical intervention, physical therapy, and occupational therapy (OT), increase the effectiveness of the treatment program 69. Family members are involved in the evaluation and treatment processes.
Appropriate caution must be taken during chronic pain syndrome treatment in patients who exhibit any of the following behaviors:
- Poor response to prior appropriate management
- Unusual, unexpected response to prior specific treatment
- Avoidance of school, work, or other social responsibility
- Severe depression
- Severe anxiety disorder
- Excessive pain behavior
- Physician shopping
- Noncompliance with treatment in the past
- Drug abuse or dependence
- Family, marital, or sexual problems
- History of physical or sexual abuse
National health professional organizations have issued guidelines for treating several chronic pain syndrome 70.
Hospitalization usually is not required for patients with chronic pain syndrome, but it depends on how invasive the treatment choice is for pain control and on the severity of the case.
Patients with chronic pain syndrome generally are treated on an outpatient basis and require a variety of health care professionals to manage their condition optimally.
A clinical practice guideline from the American College of Physicians 71 encourages the use of non-pharmacologic approaches as initial treatment for chronic low-back pain. The options they suggest include several complementary approaches—acupuncture, mindfulness-based stress reduction, tai chi, yoga, progressive relaxation, biofeedback, and spinal manipulation—as well as conventional methods such as exercise and cognitive behavioral therapy.
The American College of Rheumatology 72 mentions several complementary approaches in its guidelines for the management of osteoarthritis of the hip or knee. For osteoarthritis of the knee, the guidelines mention tai chi as one of several nondrug approaches that might be helpful. The same guidelines, however, discourage using the dietary supplements glucosamine and chondroitin for osteoarthritis of the hip or knee.
The American College of Gastroenterology 73 included probiotics/prebiotics, peppermint oil, and hypnotherapy in its evaluation of approaches for managing irritable bowel syndrome. The American College of Gastroenterology found only weak evidence that any of these approaches may be helpful.
Table 2. Chronic Pain Treatment Options
Non-Drug Options | Nociceptive | Neuropathic | Central Sensitization |
---|---|---|---|
Consider in all patients | |||
Increased Activity/Exercise (aerobic, strength, flexibility/balance) | In addition to helping generally across types of pain, specifically: | ||
Knee and hip osteoarthritis Rheumatoid arthritis Vascular claudication, | Chemotherapy induced neuropathy Diabetic peripheral neuropathy Multiple sclerosis | Fibromyalgia (for pain, aerobic exercise rather than resistance exercise) | |
Improve Sleep | All types of pain | ||
Dietary Modification | For all types of pain: Mediterranean pattern of eating | ||
Self-regulatory and psychophysiological approaches | For all types of pain: biofeedback, relaxation training, and hypnosis | ||
Consider in select patients based on diagnosis and interest | |||
Acupuncture | Osteoarthritis, chronic neck and low back pain, Headache | Post-herpetic neuralgia, chemotherapy induced polyneuropathy | Fibromyalgia |
Physical Therapy | Functional deficits or secondary pain generators that directed therapy may improve (based on functional deficits rather than diagnosis) | ||
Transcutaneous electrical stimulation (TENS) | Rheumatoid arthritis Knee osteoarthritis | Diabetic peripheral neuropathy Post herpetic neuralgia | Fibromyalgia |
Massage | Low back pain Knee osteoarthritis Neck pain Hand osteoarthritis | Fibromyalgia | |
Mindfulness Based Stress Reduction | Low back pain Rheumatoid arthritis | Fibromyalgia | |
Cognitive behavioral therapy (CBT) | Low back pain Neck pain Knee pain Shoulder pain Hip pain, Hip osteoarthritis Knee osteoarthritis Rheumatoid arthritis Systemic lupus erythematosus Temporomandibular joint pain. | Fibromyalgia | |
Acceptance and commitment therapy (ACT) | Low back pain Rheumatoid arthritis | Fibromyalgia |
Regular exercise
Extensive research on physical inactivity in patients with chronic pain has revealed that the majority experience a reduction in overall physical activity levels due to the impact of chronic pain 75, 76, 77, 78. The more debilitating the pain, the lower the engagement in physical activity 79.
People who have chronic pain are advised to stay physically active and get regular exercise 80, 81. According to the available evidence, physical activity did not cause harm in people with chronic pain 81. When you exercise, your body releases substances that have a pain-relieving effect. Exercise also improves your blood circulation and stimulates your metabolism, making sure that your bones and cartilage get enough nutrients. It has other advantages too: exercise improves your physical resilience, your ability to move and your sense of balance, which can help to prevent falls in old age. Last, but not least, it can improve your wellbeing.
The National Institute for Health and Care Excellence (NICE) osteoarthritis guidelines state that “for all people with osteoarthritis, offer therapeutic exercise tailored to their needs for example, local muscle strengthening, general aerobic fitness” 82:
- doing regular and consistent exercise, even though this may initially cause pain or discomfort, will be beneficial for their joints
- long-term adherence to an exercise plan increases its benefits by reducing pain and increasing functioning and quality of life.
People who aren’t used to exercising should start off slowly to avoid overdoing it. You can find out what type of exercise helps you by either trying out different things yourself or as part of exercise-based treatment. The aim of exercise-based treatment is to gradually increase your pain threshold again. There are special programs for a number of pain syndromes such as rheumatoid arthritis, fibromyalgia and chronic back pain, for example organized by support groups. In Germany, doctors can also prescribe something known as functional training for up to 24 months.
Diet and Weight Management
Diet represents another modifiable lifestyle factor of significant importance to patients with chronic pain 83. Poor dietary habits can be considered as adverse lifestyle factors that partly account for the observed excess mortality among people with chronic pain 84, 85. Research indicates that the dietary quality of individuals with chronic pain tends to be lower than that of those without pain 86, 87, 88. Evidence suggests that nutrition should be considered within a personalized approach to pain management 89.
Collectively, unhealthy dietary patterns, overweight and obesity are increasingly acknowledged as viable targets for therapeutic intervention in individuals with chronic pain 90, 91, 92, 93, 94. Poor dietary habits often but not always relate to overweight or obesity 95. Meta-analyses confirm a positive association between overweight, obesity and low back pain 95, 96, 97, 98, 90, 91, 99, with a higher body mass index (BMI) and fat mass correlating with increased prevalence of chronic pain 55. Furthermore, overweight and obesity contribute to more intense and debilitating chronic pain, as evidenced by dose–response relationships between pain intensity, disability, body mass index (BMI), waist circumference, percent fat and fat mass in individuals with chronic low back pain 91. In terms of socioeconomic impact, overweight and obesity correlate not only with persistent chronic pain but also with elevated healthcare-seeking rates for pain-related issues 95. Individuals who are obese or overweight and experiencing chronic pain are likely to have more health needs that necessitate a focus on behavioral lifestyle factors 92.
Studies involving overweight and obese adults with knee osteoarthritis demonstrate that a combination of diet and exercise therapy can yield moderate pain relief and improved physical function 100. In a more recent randomized controlled trial 101, three interventions were compared: exercise alone, intensive diet-induced weight loss alone and a combination of intensive diet-induced weight loss with exercise. The study revealed that in comparison to exercise alone, diet-induced weight loss led to substantial reductions in load bearing at the hip, knee and ankle joints 101. However, when diet was combined with exercise, these reductions were less pronounced, although they remained significantly superior to the results achieved with exercise alone 101. Additionally, it is worth noting that diet and exercise therapy, when integrated into standard care, have proven to be cost-effective for such patients 102.
Weight management programs should include changes in dietary and physical activity behaviors (i.e., a self-management behavioral approach to balance caloric intake and physical activity) to comply with evidence-based standards rather than strict and harsh calorie restriction diets 94. This should involve the integration of behavioral changes to support and facilitate calories balance-related behavior 103. The American College of Sports Medicine (ACSM) emphasize the combination of a moderate dietary restriction and a physical activity program is effective and yields longterm outcomes 104. Adopting a self-management approach to enhance behaviors related to calories balance can be seamlessly incorporated into the previously outlined cognition-targeted exercise therapy program. This implies that assessing daily physical activity and exercise levels will be geared toward not only weight reduction but also the simultaneous improvement of pain-related cognitions. Consequently, the exercises and daily physical activities can be aligned with the principles of cognition-targeted treatment as well as the personalized approaches for weight reduction 89.
While specific recommendations may vary based on individual needs, general guidelines for dietary interventions in chronic pain advocate for the inclusion of colorful fruits and vegetables, along with an adequate intake of high-quality fats to reduce inflammation and oxidative stress 86. Additionally, preventing deficiencies in Vitamin D, Vitamin B12 and Magnesium has the potential to contribute to pain alleviation 86, 105, 106. Lastly, ensuring sufficient fiber intake proves significant for promoting proper digestion, maintaining a healthy microbiome, managing weight and subsequently influencing chronic pain 86.
Complementary medicine
Some recent research has looked at the effects of complementary approaches on chronic pain syndrome in general rather than on specific painful conditions. A growing body of evidence suggests that some complementary approaches, such as acupuncture, hypnosis, massage, mindfulness meditation, spinal manipulation, tai chi, and yoga, may help to manage some painful conditions.
- A 2017 review 107 looked at complementary approaches with the opioid crisis in mind, to see which ones might be helpful for relieving chronic pain and reducing the need for opioid therapy to manage pain. There was evidence that acupuncture 108, yoga, relaxation techniques, tai chi, massage, and osteopathic or spinal manipulation 109 may have some benefit for chronic pain, but only for acupuncture was there evidence that the technique could reduce a patient’s need for opioids.
- Research shows that hypnosis is moderately effective in managing chronic pain, when compared to usual medical care. However, the effectiveness of hypnosis can vary from one person to another.
- A 2017 review of studies of mindfulness meditation for chronic pain showed that it is associated with a small improvement in pain symptoms.
- Studies have shown that music can reduce self-reported pain and depression symptoms in people with chronic pain 110.
- Massage therapy is manual manipulation of muscles and connective tissue to enhance physical rehabilitation and improve relaxation. Massage therapy can reduce pain scores for patients with low back pain, knee osteoarthritis, juvenile rheumatoid arthritis, chronic neck pain, and fibromyalgia 111, 112. Not yet determined are the optimal number, duration and frequency of massage sessions for treating pain.
- Transcutaneous electrical nerve stimulation (TENS). Transcutaneous electrical nerve stimulation (TENS) uses low-voltage electrical currents to relieve pain. A TENS unit is a small device that delivers the current at or near your nerves to block or change your perception of pain. Researchers are still working to find out more information. Healthcare providers use TENS to treat a range of conditions, including osteoarthritis, tendinitis and fibromyalgia. Most experts believe the electrical current helps release pain-reducing chemicals that your own body produces. Evidence is limited for the efficacy of TENS in pain management 113. However, it is relatively safe, with units relatively available and easy to use.
Although the mind and body practices studied for chronic pain have good safety records, that doesn’t mean that they’re risk-free for everyone. Your health and special circumstances (such as pregnancy) may affect the safety of these approaches. If you’re considering natural products, remember that natural doesn’t always mean safe and that some natural products may have side effects or interact with medications.
Mind and body practices, such as acupuncture, hypnosis, massage therapy, mindfulness/meditation, relaxation techniques, spinal manipulation, tai chi/qi gong, and yoga, are generally safe for healthy people if they’re performed appropriately.
- People with medical conditions and pregnant women may need to modify or avoid some mind and body practices.
- Like other forms of exercise, mind and body practices that involve movement, such as tai chi and yoga, can cause sore muscles and may involve some risk of injury.
- It’s important for practitioners and teachers of mind and body practices to be properly qualified and to follow appropriate safety precautions.
Table 3. Herbal Supplements used in Chronic Pain
Name | Proposed Indication | Proposed Effect | Side Effects/Notes |
---|---|---|---|
Arnica (Arnica montana) (topical) 42 | Low back pain – acute flares Muscle pain Osteoarthritis | Anti-inflammatory | Oral use may be toxic |
Boswellia serrata 114 | Osteoarthritis | Anti-inflammatory | |
Cannabinoids (medicinal cannabis) 115 | Neuropathic pain Osteoarthritis Headache | cannabidiol (CBD): Anti-inflammatory THC: Central nervous system mediated | Quality of evidence is low to date with only real benefit a mild one in pain reduction for neuropathic pain. cannabidiol (CBD) may play an anti-inflammatory role, with THC causing most adverse effects. Consider advising patients who are planning to use for pain management to start with low dose cannabidiol (CBD) alone. |
Cayenne pepper (Capsicum annuum) (topical) 42 | Low back pain –acute flares | Analgesic Anti-Inflammatory | Equal to placebo |
Devil’s claw (Harpagophytum procumbens) 60-100 mg (standardized hapagosides) / day in divided dosing (Harpagophutum) 115 | Low back pain – acute flares | Analgesic Anti-inflammatory | No evidence for use in other pain conditions |
Glucosamine and chondroitin 44 | Osteoarthritis | May interact with warfarin. Does not treat pain from knee or hip OA. Does not slow the progression of knee or hip OA. Does not have disease modifying effects in knee or hip OA. | |
Turmeric (Curcumin) 116 | Osteoarthritis | Anti-inflammatory | Case reports of antiplatelet effect but only clinical trial showed no impact on bleeding or INR. |
Willow bark 120 to 240 mg/day 42 | Low back pain – acute flares | Analgesic Anti-inflammatory | Contains salicin, an aspirin precursor. Avoid if allergic to ASA or NSAIDs. |
Footnotes: * Always check with your doctor for potential interactions of herbal supplements with prescription medications or other non-prescription medications or supplements.
[Source 74 ]Chronic pain medications
Some medicines can play a role in managing chronic pain. Sometimes, medicines won’t relieve all your pain symptoms. It’s important to work with a doctor or pain specialist to identify a range of strategies you can use, to reduce the way pain impacts your life.
- Acetaminophen (paracetamol) — this very effective pain-relief medicine is sometimes taken along with other medicines. Acetaminophen is thought to block the production of prostaglandins in the central nervous system. Prostaglandins are hormonelike substances that are involved in pain and inflammation. Acetaminophen is usually recommended as a first line treatment for mild to moderate pain. Acetaminophen might be taken for pain due to a skin injury, headache, or conditions that affect the muscles and bones. Acetaminophen is often prescribed to help manage osteoarthritis and back pain. Do not take more than the daily dose listed on the package. Speak to your doctor if acetaminophen (paracetamol) is not managing your pain. Acetaminophen is not as effective as nonsteroidal anti-inflammatory drugs (NSAIDs) for the treatment of knee and hip pain related to osteoarthritis.
- Non-steroidal anti-inflammatory drugs (NSAIDs) — medicines such as ibuprofen (Advil, Motrin IB, others), naproxen sodium (Aleve) and diclofenac may help manage pain. Try to take them at the lowest possible dosage for the shortest possible time, as they can have serious side effects. But if you take more than the recommended dosage, NSAIDs may cause nausea, stomach pain, stomach bleeding or ulcers. Sometimes this can happen even when you take the recommended dosage.
- Antidepressants — some medicines that are used to treat depression, such as tricyclic antidepressants (TCAs) and serotonin and norepinephrine reuptake inhibitors (SNRIs), are also sometimes prescribed by doctors to reduce pain.
- Tricyclic antidepressants (TCAs) used in the treatment of chronic pain include amitriptyline and nortriptyline (Pamelor).
- Serotonin and norepinephrine reuptake inhibitors (SNRIs), that may be prescribed to relieve chronic pain include duloxetine (Cymbalta, Drizalma Sprinkle), venlafaxine (Effexor XR, Pristiq) and milnacipran (Fetzima, Savella).
- Anticonvulsants or antiepileptic medicines — medicines that are commonly used to treat epilepsy can also help manage nerve pain. Anti-seizure medications used to treat chronic nerve pain include gabapentin (Gralise, Neurontin, Horizant) and pregabalin (Lyrica). These medications treat the burning pain of shingles, known as postherpetic neuralgia. And they treat pain related to nerve damage in the legs and feet due to diabetes, known as diabetic neuropathy. Anti-seizure medications quiet pain signals from nerve cells. They may be helpful for stabbing or shooting pain from nerve damage.
- Muscle relaxants. Sedating or non-sedating muscle relaxants are often prescribed for chronic myofascial pain, despite little or no evidence for a long-term benefit 117. Cyclobenzaprine, tizanidine, and metaxalone can cause significant sedation, while methocarbamol is less likely to do so. Benzodiazepines pose a significant risk for long-term dependence and misuse, and they substantially increase the danger of overdose when used together with opioids. Baclofen, while somewhat useful for spasticity, has little role as a muscle relaxant, poses a significant risk for dependence, and should generally be avoided.
- Topical agents. Topical NSAIDs and anesthetics are occasionally useful in nociceptive or neuropathic pain syndromes. They can be expensive and are often not covered by insurance. Topical NSAIDs benefit a minority of osteoarthritis patients. They generally are not useful in other types of pain 118, 119. Topical lidocaine patches (prescribed or over-the-counter) can be effective. Ointment is less effective and can be messy. Both are expensive and often not covered by insurance. Over the counter 4% lidocaine cream is not expensive, but only marginally effective. Capsaicin cream (1%, not 0.25%) can be modestly effective, is available without prescription, but requires care in application to avoid unwanted burning. Compounded capsaicin 8% cream is more effective, but the cost may be prohibitive. When other treatments have failed, topical nitroglycerin may have some effect for wound pain, anal fissure pain, vulvodynia, and diabetic neuropathy. Headache can complicate treatment with nitroglycerin. Avoid nitroglycerin in patients who use phosphodiesterase type 5 inhibitors (avanafil, sildenafil, tadalafil, vardenafil) for erectile dysfunction. Compounded topical 5% morphine can provide local wound analgesia and may promote healing. It is only available at compounding pharmacies and can be expensive.
- Opioids — strong pain-relief medicines, such as morphine, fentanyl (Actiq, Fentora), oxycodone (OxyContin, Roxicodone, others), codeine, hydrocodone (Hysingla ER), hydrocodone-acetaminophen, oxycodone-acetaminophen (Percocet), might be prescribed by a doctor for short periods of time for acute pain, but are not effective in chronic pain that is not caused by cancer. Deciding whether to prescribe opioids is based on an assessment of benefits and harms. Opioids are not usually recommended for most types of chronic pain 120. This is because opioids are proven to not be effective at treating chronic pain not caused by cancer. Research shows that over time your body adapts to these medications and they bring less and less pain relief. This is known as tolerance. It means that you need more of the medication to achieve the same degree of pain relief. Long-term use of opioids may lead to dependence on the medications and, eventually, addiction. The longer you use opioids, the greater the risk of becoming addicted. But even using opioids to manage pain for more than a few days increases your risk. Researchers have found that the odds of being on opioids a year after starting a short course increases after only five days on the medication.
Many medications commonly used to treat chronic pain carry potential risks, side effects, and possible complications. Acetaminophen (paracetamol) is a standard pharmacological therapy for patients with chronic pain, taken either as a single agent or in combination with an opioid. However, liver toxicity can occur, especially with doses exceeding 4 grams per day, making it the most common cause of acute liver failure in the US 121, 122. Furthermore, liver toxicity can occur at therapeutic doses for patients diagnosed with chronic liver disease 123.
Frequently used add-on (adjunct) medications such as gabapentin or pregabalin can cause sedation, swelling, mood changes, confusion, and respiratory depression in older patients who require additional pain relievers 124. Add-on (adjunct) medications such as gabapentin or pregabalin require caution in older patients with painful diabetic neuropathy. Moreover, combining gabapentin or pregabalin with opioid analgesics has been shown to increase death rate of patients 125.
Duloxetine can cause mood changes, headaches, nausea, and other possible side effects and should be avoided in patients with a history of kidney or liver disease. Patients with these conditions should be closely monitored if duloxetine is prescribed.
Feared complications of opioid therapy include addiction, overdose resulting in respiratory compromise, and occasionally, death. However, opioid-induced hyperalgesia is also a significant concern, where patients become more sensitive to painful stimuli while on chronic opioids 126. The long-term risks and side effects of opioids include constipation, tolerance, dependence, nausea, dyspepsia, arrhythmia (methadone treatment QT prolongation), and opioid-induced endocrine dysfunction, which can result in amenorrhea, impotence, gynecomastia, and decreased energy and libido 1. Additionally, there appears to be a dose-dependent risk of opioid overdose with increasing daily milligram morphine equivalents 1.
Table 4. Chronic pain medications
Medication | May Benefit | Potential Co-treatment Of | Harms | Cost | Comments |
---|---|---|---|---|---|
Acetaminophen (paracetamol) | Nociceptive | Headaches | May exacerbate chronic daily headaches | Low | May be synergistic when combined with NSAIDs |
Non-steroidal anti-inflammatory drugs (NSAIDs) | Nociceptive | Headaches | Gastrointestinal bleeding, acute kidney injury, chronic kidney disease, increased risk for coronary artery events | Low | May increase blood pressure; edema. COX-2 inhibitor somewhat decreases risk of gastrointestinal bleeding |
Serotonin and norepinephrine reuptake inhibitors (SNRIs) antidepressants Duloxetine, venlafaxine, milnacipran | Central pain sensitization (Type 1) pains, neuropathic pain, non-specific low back pain, functional abdominal pain | Anxiety Depression | Weight gain, urinary retention, withdrawal symptoms (taper down to discontinue) | Low/Moderate | Duloxetine FDA-approved for diabetic neuropathy, fibromyalgia Duloxetine more effective than venlafaxine |
Anticonvulsants Gabapentin Pregabalin Topiramate | Neuropathic pain, fibromyalgia Neuropathic pain | Gabapentin: menopausal hot flushes Migraine prophylaxis | Weight gain, edema, fatigue Cognition and speech problems | Gabapentin: Low Pregabalin: Low Topiramate: Moderate | Gabapentin: not effective in low back pain Pregabalin: FDA-approved for diabetic neuropathy, fibromyalgia |
Tricyclic antidepressants (TCAs) | Central, neuropathic | Anxiety, depression, insomnia, migraine prophylaxis, smoking cessation | Fatigue, weight gain, constipation | Low | Give in early evening when sleep initiation is an issue |
Muscle relaxants Cyclobenzaprine, methocarbamol, tizanidine, Benzodiazepines (BZD), carisoprodol, Baclofen | Muscle spasms Spasticity | Fatigue Sedation, dependence Same as benzodiazepines | Low/High | Not effective for acute or chronic back pain. Benzodiazepines, carisoprodol (Soma): neither indicated nor effective – high risk for dependence Same as benzodiazepines | |
Topical Agents Non-steroidal anti-inflammatory drugs (NSAIDs)Lidocaine ointment or patch Capsaicin cream Nitroglycerin | Osteoarthritic (OA) joints OA joints, focal neuropathic pain Same as lidocaine Wound, anal fissure pain, vulvodynia, diabetic neuropathy | Headaches | High/Very High High/Very High Low Low | Ointment is messy. Patches often not covered by insurance Do not use nitroglycerin in patients using PDE-5 erectile dysfunction medications |
Managing your chronic back pain
Managing chronic pain means finding ways to make your back pain tolerable so you can live your life 127. You may not be able to get rid of your pain completely, but you can change some things that worsen your pain. These things are called stressors. Some of them may be physical, like the chair you sit in at work. Some may be emotional, like a difficult relationship.
Reducing stress can improve your physical and emotional health. It is not always easy to reduce stress, but it’s easier if you are able to ask your friends and family for help.
Make a List
First, make a list of what makes your back pain better and what makes it worse.
Then try to make changes in your home and work to decrease the causes of your pain. For example, if bending to pick up heavy pots sends shooting pain down your back, rearrange your kitchen so that the pots are hanging from above or are stored at waist height.
If your back pain is worse at work, talk to your boss. It may be that your workstation isn’t set up correctly.
- If you sit at a computer, make sure that your chair has a straight back with an adjustable seat and back, armrests, and a swivel seat.
- Ask about having an occupational therapist assess your workspace or movements to see if changes such as a new chair or cushioned mat under your feet would help.
- Try not to stand for long periods. If you must stand at work, rest one foot on a stool, then the other foot. Keep switching the load of your body’s weight between your feet during the day.
Long car rides and getting in and out of the car can be hard on your back. Here are some tips:
- Adjust your car seat to make it easier to enter, sit in, and get out of your car.
- Bring your seat as far forward as possible to avoid leaning forward when you are driving.
- If you drive long distances, stop and walk around every hour.
- DO NOT lift heavy objects right after a long car ride.
These changes around your home could help relieve your back pain:
- Raise your foot up to the edge of a chair or stool to put your socks and shoes on instead of bending over. Also consider wearing shorter socks. They are quicker and easier to put on.
- Use a raised toilet seat or install handrail next to the toilet to help take pressure off your back when you sit on and get up from the toilet. Also make sure the toilet paper is easy to reach.
- DO NOT wear high-heeled shoes. If you must wear them sometimes, consider wearing comfortable shoes with flat soles to and from the event or until you must put on high-heels.
- Wear shoes with cushioned soles.
- Rest your feet on a low stool while you’re sitting so that your knees are higher than your hips.
Pain Relievers and Medicines
Pain relievers are medicines that reduce or relieve headaches, sore muscles, arthritis, or other aches and pains. There are many different pain medicines, and each one has advantages and risks. Some types of pain respond better to certain medicines than others. Each person may also have a slightly different response to a pain reliever.
Over-the-counter (OTC) medicines are good for many types of pain. There are two main types of OTC pain medicines: acetaminophen (Tylenol) and nonsteroidal anti-inflammatory drugs (NSAIDs). Aspirin, naproxen (Aleve), and ibuprofen (Advil, Motrin) are examples of OTC NSAIDs. If you take OTC pain medicines, make sure you do read and follow the instructions on the box.
If OTC medicines don’t relieve your pain, your doctor may prescribe something stronger or long-acting medicines for constant pain. Many NSAIDs are also available at higher prescription doses. The most powerful pain relievers are opioids. They are very effective, but they can sometimes have serious side effects. There is also a risk of addiction. Because of the risks, you must use them only under a doctor’s supervision.
There are many things you can do to help ease pain. Pain relievers are just one part of a pain treatment plan.
Your doctor might recommend a prescription pain reliever. Be sure to follow his or her instructions for how to take the medicine. Many prescription pain relievers are opioids. Opioids can be very effective when taken as directed. But many people who misuse opioids become addicted. Opioid addiction is a very serious health issue that can lead to death.
Rely on Friends and Family
It is important to have strong relationships with family and friends you can depend on when your back pain makes it hard to get through the day.
Take time to build strong friendships at work and outside of work by using caring words and being kind. Give sincere compliments to the people around you. Respect those around you and treat them the way you like to be treated.
If a relationship is causing stress, consider working with a counselor to find ways to resolve conflict and strengthen the relationship
Establish Life Routines
Set up good life habits and routines such as:
- Exercise a little every day. Walking is a good way to keep your heart healthy and your muscles strong. If walking is too hard for you, work with a physical therapist to develop an exercise plan that you can do and maintain.
- Eat foods that are low in fats and sugar. Healthy foods make your body feel better, and they decrease your risk of being overweight, which can cause back pain.
- Reduce demands on your time. Learn how to say yes to things that are important and no to those that are not.
- Prevent pain from starting. Figure out what causes your back pain, and find other ways to get the job done.
- Take medicines as needed.
- Make time for activities that make you feel relaxed and calm.
- Give yourself extra time to get things done or to get where you need to go.
- Do things that make you laugh. Laughter can really help reduce stress.
- Getting regular sleep at night and not taking daytime naps should help.
- Stopping smoking also helps, because the nicotine in cigarettes can make some medicines less effective. Smokers also tend to have more pain than nonsmokers 40.
What is Complex Regional Pain Syndrome?
Complex regional pain syndrome (CRPS), also known as reflex sympathetic dystrophy or causalgia, is a chronic (lasting greater than 3 months) pain condition that is regional pain (not in a specific nerve territory or dermatome) most often affects one limb (arm, leg, hand, or foot) usually after an injury, a surgery, a stroke or a heart attack 128, 129, 130, 131, 132, 133, 134, 135, 136, 137. The pain is out of proportion to the severity or duration to the usual course of pain after such tissue trauma 138. For example, complex regional pain syndrome is a syndrome that can occur after simple injuries such as a sprained ankle or a broken arm. Complex regional pain syndrome (CRPS) is characterized by signs indicating autonomic and neuro-inflammatory changes in the affected body region varying between patients and over time. Complex regional pain syndrome is characterized by prolonged or excessive pain and changes in skin color, temperature, stiffness, weakness and/or swelling in the affected area. Often, complex regional pain syndrome (CRPS) is accompanied by significant emotional distress or functional disability. Complex regional pain syndrome is believed to be caused by damage to or malfunction of, the peripheral nervous system (nerves that are located outside of your brain and spinal cord) and central nervous system (brain and spinal cord) 139, 140, 131, 141. Complex regional pain syndrome is almost three or four times more common in women than in men, and peaks in onset between the ages of 50 and 70 years 142. Given the changes in diagnostic criteria and the evolving understanding of complex regional pain syndrome process, studies vary on the actual incidence of complex regional pain syndrome. The overall incidence rates of complex regional pain syndrome can range anywhere from approximately 5 to 29 per 100,000 people each year, with the highest incidence typically occurring in females between 61 and 70 years of age 143, 144, 145. Based on the epidemiological studies available, complex regional pain syndrome appears to be more common in patients of North European ancestry, although it does appear to occur frequently in those of South Korean ancestry as well 144, 145. In the United States, it is estimated that there are at least 50,000 cases of complex regional pain syndrome type 1 each year 146. Similarly, with the variation in complex regional pain syndrome diagnostic criteria over the years the incidence among patients with fractures is anywhere from 0.05% to 0.2% in older studies and 3% to 7% in more recent studies 147. The upper extremity is affected more frequently than the lower extremity 144.
Signs and symptoms of complex regional pain syndrome include:
- Continuous burning or throbbing pain, usually in your arm, leg, hand or foot
- Sensitivity to touch or cold
- Swelling of the painful area
- Changes in skin temperature — alternating between sweaty and cold
- Changes in skin color, ranging from white and mottled to red or blue
- Changes in skin texture, which may become tender, thin or shiny in the affected area
- Changes in hair and nail growth
- Joint stiffness, swelling and damage
- Muscle spasms, tremors, weakness and loss (atrophy)
- Decreased ability to move the affected body part
Symptoms may change over time and vary from person to person. Pain, swelling, redness, noticeable changes in temperature and hypersensitivity particularly to cold and touch usually occur first.
Over time, the affected limb can become cold and pale. It may undergo skin and nail changes as well as muscle spasms and tightening. Once these changes occur, the condition is often irreversible.
Complex regional pain syndrome occasionally may spread from its source to elsewhere in your body, such as the opposite limb.
In some people, signs and symptoms of complex regional pain syndrome go away on their own. In others, signs and symptoms may persist for months to years. Treatment is likely to be most effective when started early in the course of the illness.
Complex regional pain syndrome is divided into two types 148:
- Complex regional pain syndrome type 1: Individuals without a confirmed nerve injury are classified as having Complex regional pain syndrome type 1 (previously known as reflex sympathetic dystrophy syndrome).
- Complex regional pain syndrome type 2: Complex regional pain syndrome type 2 (previously known as causalgia) is when there is an associated, confirmed nerve injury.
As some research has identified evidence of nerve injury in complex regional pain syndrome type 1, it is unclear if this disorder will always be divided into two types 149, 150, 151, 152, 153. Nonetheless, the treatment is similar. Both subtypes of complex regional pain syndrome are characterized by severe pain that is disproportionate to the inciting event, most commonly affecting the hand or foot but which can spread to other body regions 154. Additionally complex regional pain syndrome presents with some or all of the following symptoms in the affected body parts: sensory disturbances, temperature changes, abnormal patterns of sweating, swelling and edema, reduced joint range of motion, movement abnormalities such as weakness, tremor or dystonia, trophic changes such as skin atrophy or altered hair and nail growth, and localized osteoporotic changes 155; and alterations in body perception or schema 156.
Complex regional pain syndrome is often described in terms of “warm” changes and “cold” changes, although they are not separate diagnostic categories. Warm complex regional pain syndrome is associated with a warm, redness of the skin, swelling of extremity and tends to be correlated with inflammatory changes earlier in the disease course; one study described these changes within the first eight months 157. Cold complex regional pain syndrome is associated with a cold, dusky, sweaty extremity and is more often associated with chronic complex regional pain syndrome; in one study, cold symptoms lasted for more than nine months 157. The same extremity can have both warm and cold symptoms, and the disease pattern and clinical course vary among patients 158, 159, 160.
Complex regional pain syndrome symptoms vary in severity and duration, although some cases are mild and eventually go away. In more severe cases, individuals may not recover and may have long-term disability.
Although it is more common in women, complex regional pain syndrome can occur in anyone at any age, with a peak at age 40. Complex regional pain syndrome is rare in the elderly. Complex regional pain syndrome is uncommon in children under age 10.
At the consensus meeting of the International Association for the Study of Pain (IASP) in Budapest in 2004, the Budapest criteria for the diagnosis of complex regional pain syndrome were developed 161, 159. In an effort to enhance specificity, the Budapest clinical diagnostic criteria for complex regional pain syndrome included both symptoms and signs of the disease. The Budapest criteria is 99% sensitive and 68% specific for complex regional pain syndrome, with increased sensitivity as other diagnoses are ruled out 161, 162. The Budapest criteria were formally accepted by the International Association for the Study of Pain (IASP) in 2012 after more robust validation testing 163. It should be noted that a 4 of 4 symptom decision rule is recommended when applying the Budapest criteria for research purposes 162. The Budapest criteria, are listed in Table 1 below.
Additional clinical findings in complex regional pain syndrome 164:
- Allodynia
- pain on light movement/touch/pressure
(e.g., draught, brush, finger [pressure less than 100 g/cm² or load less than 500 g]) - pain from vibration (e.g. tuning fork on a bony prominence), cold, heat
- pain on light movement/touch/pressure
- Hyperalgesia
- periarticular pressure hyperalgesia distal to the site of injury/surgery
- solitary sharp stimulus (pinprick) is felt stronger/longer
- Disturbance of body perception/interoception
- attention: exerted, hypervigilant focussing or disregarding (neglect-like/dissociation)
- shape, size, weight, position, posture, laterality, symmetry, ownership, e.g., as if limbs were detached, foreign, dead (“as if wooden”, “like part of a dead body”)
- attribution of meaning or affective evaluation, e.g., threatening, disgusting, or hated limb; caution: desire for amputation
- Functional disturbances of voluntary motor function
- protective posture, slowed, reduced or non-movement
- reduced experience of motor action, control, intention (agency)
- weakness, disturbances of fine motor skills, coordination or movement
- sometimes several types of disturbance inone and the same limb, e.g.
- myoclonus or tremor (with “entrainment”, i.e. unconscious synchronization with rhythmic movements of the unaffected limb)
- dystonia (usually in fixed flexion/supination)
- increase during manipulation, decrease when distracted, no decrease with sensory tricks
- Trophic, vasomotor and sudomotor disturbances
- non-pitting edema (circumference measurements where possible)
- skin color usually reddish-blue, with time more waxy-pale
- skin temperature about 1–2 degrees difference to the contralateral side
- hair growth: usually increased, thick, dark, sometimes reduced, finer hair
- nail growth: usually harder, brittle, faster or slower nail growth
- increased, with time reduced, sweating
- trophic skin lesions or pressure lesions
- Tendency to spread and become generalized
- at times tendency to spread proximally or to other limbs
- other pain sites; contralateral and generalized reduced pain threshold/body image disturbance
- sensitivity to noise, light, smell, touch, at times with autonomic arousal
- cardiovascular dysregulation, increased heart rate, reduced heart rate variability
- micturition and digestive problems
- Affective, cognitive, general signs and symptoms; behavioral changes
- fear, particularly of touch, movement, and disease progression
- avoidance of sensory stimuli from clothes, wind, movement, other people, etc.
- signs and symptoms of trauma-related disorders (helplessness, flashbacks, dissociation, depersonalization, avoidance, hypervigilance/hyperarousal)
- mood fluctuations, depressiveness, alexithymia (reduced awareness of one’s own emotions and those of others), withdrawal
- fatigue, sleeping problems, and cognitive disturbances
Table 1. The Budapest clinical diagnostic criteria for complex regional pain syndrome
1. Continuing pain, which is disproportionate to any inciting event |
2. Must report at least one symptom in all four following categories
|
3. Must display at least one sign at time of evaluation in two or more of the following categories
|
4. There is no other diagnosis that better explains the signs and symptoms |
Figure 1. Complex regional pain syndrome pathophysiology
Footnotes: Pathophysiological connections in the complex regional pain syndrome. An insulting event triggers classic and neurogenic inflammation, which are responsible for the inflammatory signs at least of the warm phase and, perhaps, contribute to the autonomic dysfunction. Autonomic dysfunction is also responsible for several aspects of the syndrome, while interventions targeting it have been beneficial, mainly in case series. Central nervous system (brain and spinal cord) plasticity and psychological factors likely complete the process. However, the exact mechanisms eliciting the central nervous system (brain and spinal cord) changes, linking to the psychological factors and ultimately implicating both of these factors to the clinical presentation of the syndrome are still obscure.
[Source 148 ]Complex regional pain syndrome causes
It is unclear why some individuals develop complex regional pain syndrome while others with similar trauma do not 166, 167, 168. In more than 90 percent of cases, complex regional pain syndrome is triggered by a clear history of trauma or injury 169, 170, 171. The most common triggers are fractures, sprains/strains, soft tissue injury (such as burns, cuts, or bruises), limb immobilization (such as being in a cast), surgery, or even minor medical procedures such as needle stick. Although complex regional pain syndrome can develop after any injury, the most common initiating events are fractures, surgery, crush injuries, and sprains 172. Complex regional pain syndrome represents an abnormal response that magnifies the effects of the injury. Some people respond excessively to a trigger that causes no problem for other people, such as what is observed in people who have food allergies.
Peripheral nerve abnormalities found in individuals with complex regional pain syndrome usually involve the small unmyelinated and thinly myelinated sensory nerve fibers (axons) that carry pain messages and signals to blood vessels. Myelin is a mixture of proteins and fat-like substances that surround and insulate some nerve fibers. Because small fibers in the nerves communicate with blood vessels, injuries to the fibers may trigger the many different symptoms of complex regional pain syndrome. Molecules secreted from the ends of hyperactive small nerve fibers are thought to contribute to inflammation and blood vessel abnormalities. These peripheral nerve abnormalities in turn trigger damage in the spinal cord and brain.
Blood vessels in the affected limb may dilate (open wider) or leak fluid into the surrounding tissue, causing red, swollen skin. The dilation and constriction of small blood vessels is controlled by small nerve fiber axons as well as chemical messengers in the blood. The underlying muscles and deeper tissues can become starved of oxygen and nutrients, which causes muscle and joint pain as well as damage. The blood vessels may over-constrict (clamp down), causing cold, white, or bluish skin.
Complex regional pain syndrome also affects the immune system. High levels of inflammatory chemicals (cytokines) have been found in the tissues of people with complex regional pain syndrome. These contribute to the redness, swelling, and warmth reported by many affected individuals. Complex regional pain syndrome is more common in individuals with other inflammatory and autoimmune conditions such as asthma.
Limited data suggest that complex regional pain syndrome also may be influenced by genetics. Rare family clusters of complex regional pain syndrome have been reported. Familial complex regional pain syndrome may be more severe with earlier onset, greater dystonia, and more than one limb being affected. When comparing patients with sporadic complex regional pain syndrome to families where two or more patients are affected, familial complex regional pain syndrome patients had a younger age at onset and more often had multiple affected extremities and dystonia 173. A recent study demonstrated that a single nucleotide polymorphism in four genes, ANO10, P2RX7, PRKAG1, and SLC12A9, is associated with developing complex regional pain syndrome type 1 with males typically expressing these rare alleles 174. Other genetic studies have demonstrated associations between complex regional pain syndrome and several major histocompatibility complex alleles, including human leukocyte antigen (HLA)-DR6, HLA-DR13, HLA-DR2, HLA-DQ1, HLA-B62, and HLA-DQ8 175. Genome-wide expression profiling of the whole blood has shown that HLA-A29.1, matrix metallopeptidase 9 (MMP9), alanyl aminopeptidase (AAP), histidine decarboxylase (HDC), granulocyte colony-stimulating factor 3 receptor (G-CSF3-R), and signal transducer and activator of transcription 3 genes (STAT-3) were highly expressed when compared to unaffected controls 176. Further genetic and transcriptomic studies hold the promise to predict patient predispositions to complex regional pain syndrome, unravel the molecular mechanisms of complex regional pain syndrome pathogenesis, and discover novel therapeutic targets for complex regional pain syndrome 177.
Occasionally complex regional pain syndrome develops without any known injury. In these cases, an infection, a blood vessel problem, or entrapment of the nerves may have caused an internal injury. A physician will perform a thorough examination in order to identify a cause.
In many cases, complex regional pain syndrome results from a variety of causes. In such instances, treatments are directed at all of the contributing factors.
Risk factors for developing complex regional pain syndrome
Potential risk factors for developing complex regional pain syndrome 164:
- Female sex, in particular postmenopausal women (women of middle age also sustain fractures three times more often than men)
- Severe injuries, in particular crush injuries, displaced, juxta-articular or intra-articular fractures (especially distal radius fractures); ankle injuries; nerve injuries; prolonged anesthesia/ischemia, manipulation procedures such as secondary fracture reductions; restricted range of motion and functional impairment, prolonged immobilization
- High pain intensity initially or directly after surgery, sensory disturbances and low skin temperature (for example, persistent pain ≥5 on a numerical rating scale of 10 points for more than a week after conservative treatment of a distal radius fracture)
- Chronic or widespread pain and hypersensitivity, such as migraine, fibromyalgia, back/period pain, neuropathies, osteoporosis, osteoarthritis, rheumatoid arthritis; also, asthma/allergies
- High anxiety level, depressiveness, passivity, avoidance tendency, pain catastrophizing, or manifest disorders associated with anxiety and trauma/stress; previous critical life events; desire for compensation and litigation.
Extremity injuries, such as fractures or sprains, surgery, and carpal tunnel syndrome have been the most commonly reported inciting events leading to complex regional pain syndrome 169, 171, 178. In a recent analysis of 1043 patients with complex regional pain syndrome, the most common primary causes were fractures (42%), blunt traumatic injuries excluding fractures (e.g., sprains) (21%), surgery (12%), and carpal tunnel syndrome (7%), while 7% had no clear precipitating event 169. However, the overall prevalence of complex regional pain syndrome associated with these inciting events remains relatively low. Crijns et al. 179 found that out of 59,765 patients treated for distal radius fractures, 0.19% were diagnosed with complex regional pain syndrome. A similar more recent study noted a 0.64% incidence of complex regional pain syndrome after 172,194 patients receiving surgical treatment for distal radius fractures 171. Complex regional pain syndrome had previously been reported as a common complication of Dupuytren contracture, but a very recent analysis of a nationwide database found the overall incidence to be 0.31% 180.
Multiple studies have demonstrated complex regional pain syndrome to be much more prevalent in women, with estimates of 2–4 times the rate in men 169, 171, 181, 182, 183. Van Velzen et al. 183 further investigated these sex-related differences, finding that male complex regional pain syndrome patients were more likely to suffer from depression and kinesiophobia, and use passive pain coping strategies. Fibromyalgia is strongly and independently associated with a diagnosis of complex regional pain syndrome, increasing the risk up to 2.5 times that of controls 179, 184. Other musculoskeletal conditions, such as rheumatoid arthritis, may also increase the risk 185. Although complex regional pain syndrome and other autonomic disorders have been anecdotally reported as adverse events after human papillomavirus (HPV) vaccination, review of published reports and a small population-based case series found no statistically significant correlation 186, 187, 188.
Complex regional pain syndrome prevention
These steps might help you reduce the risk of developing complex regional pain syndrome:
- Taking vitamin C after a wrist fracture. Studies have shown that people who took a daily minimum dose of 500 milligrams (mg) of vitamin C after a wrist fracture had a lower risk of complex regional pain syndrome compared with those who didn’t take vitamin C.
- Early mobilization after a stroke. Some research suggests that people who get out of bed and walk around soon after a stroke (early mobilization) lower their risk of complex regional pain syndrome.
Complex regional pain syndrome symptoms
The key symptom of complex regional pain syndrome is prolonged severe pain that may be constant. It has been described as “burning,” “pins and needles” sensation, or as if someone were squeezing the affected limb. The pain may spread to the entire arm or leg, even though the injury might have only involved a finger or toe. In rare cases, pain can sometimes even travel to the opposite extremity. There is often increased sensitivity in the affected area, known as allodynia, in which normal contact with the skin is experienced as very painful.
People with complex regional pain syndrome also experience changes in skin temperature, skin color, or swelling of the affected limb. This is due to abnormal microcirculation caused by damage to the nerves controlling blood flow and temperature. As a result, an affected arm or leg may feel warmer or cooler compared to the opposite limb. The skin on the affected limb may change color, becoming blotchy, blue, purple, pale, or red.
Other common features of Complex regional pain syndrome include:
- changes in skin texture on the affected area; it may appear shiny and thin
- abnormal sweating pattern in the affected area or surrounding areas
- changes in nail and hair growth patterns
- stiffness in affected joints
- problems coordinating muscle movement, with decreased ability to move the affected body part
- abnormal movement in the affected limb, most often fixed abnormal posture (called dystonia), but also tremors in or jerking of the limb.
Complex regional pain syndrome complications
If complex regional pain syndrome isn’t diagnosed and treated early, the disease may progress to more disabling signs and symptoms. These may include:
- Tissue wasting (atrophy). If you avoid moving an arm or a leg because of pain or if you have trouble moving a limb because of stiffness, your skin, bones and muscles may begin to deteriorate and weaken.
- Muscle tightening (contracture). You also may experience tightening of your muscles. This may lead to a condition in which your hand and fingers or your foot and toes contract into a fixed position.
Complex regional pain syndrome diagnosis
Currently there is no specific test that can confirm complex regional pain syndrome and complex regional pain syndrome remains a clinical diagnosis of exclusion based on the history, symptoms and signs assisted with the application of the Budapest criteria 131, 148, 128. Complex regional pain syndrome diagnosis is based on a person’s medical history, and signs and symptoms that match the definition. Since other conditions can cause similar symptoms, careful examination is important. As most people improve gradually over time, the diagnosis may be more difficult later in the course of the disorder.
Testing also may be used to help rule out other conditions, such as arthritis, Lyme disease, generalized muscle diseases, a clotted vein, or small fiber polyneuropathies, because these require different treatment. The distinguishing feature of complex regional pain syndrome is that of an injury to the affected area. Such individuals should be carefully assessed so that an alternative treatable disorder is not overlooked.
Magnetic resonance imaging (MRI) or triplephase bone scans may be requested to help confirm a diagnosis. While complex regional pain syndrome is often associated with excess bone resorption, a process in which certain cells break down the bone and release calcium into the blood, this finding may be observed in other illnesses as well.
- Bone scan with technetium Tc 99m. This procedure might help find bone changes. A radioactive substance injected into one of your veins allows your bones to be seen with a special camera.
- Sympathetic nervous system tests. These tests look for disturbances in your sympathetic nervous system. For example, thermography measures the skin temperature and blood flow of your affected and unaffected limbs. Other tests can measure the amount of sweat on both limbs. Uneven results can indicate complex regional pain syndrome.
- Infrared thermometry or thermography has the highest specificity of any objective test for complex regional pain syndrome 162. In earlier phases of the disease, the affected limb typically shows higher temperatures compared to the opposite limb 189. This observation has been shown to reverse with disease progression. Sensitivity for complex regional pain syndrome has been reported as low as 45% 190. Thermography requires specialized equipment and accuracy depends on the ability to maintain thermoregulation during measurements 162, 191. The utility of other autonomic function tests including, quantitative sudomotor axon reflex testing and thermoregulatory sweat testing, have similar limitations in that they require specialized equipment, knowledgeable operators, and they have a lower specificity for complex regional pain syndrome 162, 190.
- X-rays. Loss of minerals from your bones may show up on an X-ray in later stages of the disease. However, X-ray has low reported sensitivity and specificity for complex regional pain syndrome 192, 190. X-ray greatest utility is exclusion of other musculoskeletal injuries, particularly in early diagnosis of a unilateral extremity presentation of the disease.
- Magnetic resonance imaging (MRI). Images captured by an MRI test may show a number of tissue changes. Magnetic resonance imaging (MRI) is especially useful in the exclusion of musculoskeletal disorders, particularly osteonecrosis 190. MRI has been shown to be less sensitive for complex regional pain syndrome diagnosis than bone scan in comparative studies 193.
- Doppler flow studies. Doppler flow studies have been suggested to assess vascular reflexes, especially in patients with symptom duration of four months or less 162. Doppler flow study can also be used for exclusion of contributing vascular pathologies, particularly venous thrombosis.
Complex regional pain syndrome treatment
There is some evidence that early (within the first few months of symptom onset) aggressive treatment of pain may prevent complex regional pain syndrome symptoms 194. Currently there are not enough high quality studies to guide clear treatment decisions in complex regional pain syndrome 195. Clinical guidelines, which represent the available evidence and the views of clinical experts in complex regional pain syndrome suggest a number of treatments that may be helpful 196. Treatment options include referral for physical and occupational therapy, a short course of oral corticosteroids, bisphosphonates, and transdermal lidocaine patches 197. Evidence suggests that bisphosphonates used for the first six months of symptoms and a short course (two to four weeks) of oral corticosteroids significantly improve pain in patients with complex regional pain syndrome 159. Often, a combination of different treatments, tailored to your specific case, is necessary 198.
The goal of treatment is to improve function and decrease pain. Complex regional pain syndrome is most likely to respond to a comprehensive, integrated, multi-disciplinary treatment approach that involves medical, psychological, and physical and occupational therapy components 159.
Rehabilitation and physical therapy and occupational therapy
An exercise program to keep the painful limb or body part moving can improve blood flow and lessen the circulatory symptoms 199. Additionally, exercise can help improve the affected limb’s flexibility, strength, and function. Rehabilitating the affected limb also can help to prevent or reverse the secondary brain changes that are associated with chronic pain. Occupational therapy can help the individual learn new ways to work and perform daily tasks.
The challenge with implementing physical and occupational therapy is the broad range of interventions available to therapists and the persistent lack of understanding regarding the underlying pathophysiology of complex regional pain syndrome 128. Regardless, a number of case reports have been published showing efficacy in a wide range of interventions, ranging from strain-counterstrain (SCS), neural mobilization techniques, and thrust manipulation 200, 201, 202. These techniques often rely on some form of neuromodulation, either through decrease in proprioceptive hyperactivity as seen in strain-counterstrain (SCS) or activation of descending pain pathways and inhibitory mechanisms in spinal manual therapy. Additionally, there is evidence of analgesic effects achieved through the release of neurotensin and oxytocin, providing patients with short-term analgesia that may subsequently enhance their ability to participate in other therapeutic modalities they may previously not have tolerated 203, 204. Additionally, there is likely a role involving catecholamines in the pathophysiological pathway of complex regional pain syndrome.
Psychotherapy
Complex regional pain syndrome and other painful and disabling conditions often are associated with profound psychological symptoms for affected individuals and their families. People with complex regional pain syndrome may develop depression, anxiety, or post-traumatic stress disorder, all of which heighten the perception of pain and make rehabilitation efforts more difficult. Treating these secondary conditions is important for helping people cope and recover from complex regional pain syndrome. Furthermore, complex regional pain syndrome patient population carries significant risk factors for suicidal ideation, including: severe pain, depressive symptoms and functional impairment. The link between chronic pain and suicidal ideation has been well established, and given the additional autonomic dysfunction associated with complex regional pain syndrome, these patients carry a significant risk for suicidality 205, 206, 207, 208.
Although the evidence is lacking in clinical trial evidence, case series have shown promise in the benefits of various forms of psychotherapy intervention, especially when combined with other non-invasive modalities 128. Additionally, adaptation of the treatment regimen to the evolution of a patient’s symptoms is critical 209. Of particular interest is the study conducted by Fialka et al. 210 who found a decrease in limb temperature in the psychotherapy arm compared to the physical therapy only group. Although the study was limited by its lack of power, it lends further support to the role psychological stress can play in the progression of complex regional pain syndrome 211, 210.
Complex regional pain syndrome medications
Several different classes of medication have been reported to be effective in treating complex regional pain syndrome, particularly when used early in the course of the disease. However, no drug is approved by the U.S. Food and Drug Administration specifically for complex regional pain syndrome, and no single drug or combination of drugs is guaranteed to be effective in every person.
Drugs to treat complex regional pain syndrome include:
- bisphosphonates, such as high dose alendronate or intravenous pamidronate
- non-steroidal anti-inflammatory drugs (NSAIDs) to treat moderate pain, including over-the-counter aspirin, ibuprofen, and naproxin
- corticosteroids that treat inflammation/swelling and edema, such as prednisolone and methylprednisolone (used mostly in the early stages of complex regional pain syndrome)
- drugs initially developed to treat seizures or depression but now shown to be effective for neuropathic pain, such as gabapentin, pregabalin, amitriptyline, nortriptyline, and duloxetine
- botulinum toxin injections
- opioids such as oxycontin, morphine, hydrocodone, fentanyl, and Vicodin. These drugs must be prescribed and monitored under close supervision of a physician, as these drugs may be addictive.
- N-methyl-D-aspartate (NMDA) receptor antagonists such as dextromethorphan and ketamine, and
- topical local anesthetic creams and patches containing numbing agents such as lidocaine.
All drugs or combination of drugs can have various side effects such as drowsiness, dizziness, increased heartbeat, and impaired memory. Inform a healthcare professional of any changes once drug therapy begins.
Non-steroidal anti-inflammatory drugs (NSAIDs)
Non-steroidal anti-inflammatory drugs (NSAIDs) are cyclooxygenase-2 (COX-2) inhibitors, preventing the synthesis of prostaglandins. Prostaglandins are inflammatory mediators and induce hyperalgesia; therefore, downregulating these molecules may disrupt the process of spinal transmission of nociceptive signals and reduce the acute-pain stage of complex regional pain syndrome 212. However, the evidence remains poor regarding the efficacy of non-steroidal anti-inflammatory drugs (NSAIDs) although there are case reports showing potential benefits 213, 214.
Steroids
Steroids are used to target the inflammatory component of the proposed complex regional pain syndrome pathophysiology mechanism. There have been several studies assessing the efficacy of steroids, at various doses, various time-intervals and in patients with varying degrees of complex regional pain syndrome duration (acute vs chronic) 215, 216, 217. Given the noteworthy and potentially significant side effects associated with prolonged steroid use, care needs to be exercised by the clinician when considering these medications. One trial in particular was notable for the significant benefits of a tapered prednisone regimen in patients with an average duration of 1.9 months since onset of complex regional pain syndrome in reducing Visual Analogue Scale (VAS) pain scores 218.
Gabapentin
Gabapentin, originally used as a muscle relaxer and anti-spasmodic medication, has been found to be an effective adjunct anticonvulsant with neural pain control potential 219. Van de Vusse et al. 220 performed a randomized double blind placebo controlled crossover study and found that while gabapentin only had a mild effect on pain in complex regional pain syndrome 1, it had a significant impact on reducing the sensory deficit in the affected limb. These results were similar in the pediatric population, as Brown et al. 221 found that gabapentin significantly reduced pain intensity scores while improving sleep. This study found that both the antidepressant amitriptyline and the anticonvulsant gabapentin were analogous in their effectiveness in reducing pain in complex regional pain syndrome 1. A limitation of these trials was the low number of participants in the trials. While commonly utilized in the treatment of complex regional pain syndrome, there are ultimately few randomized control trials with solid evidence to prove the efficacy of gabapentin, suggesting that there needs to be further research into gabapentin with respect to complex regional pain syndrome 199.
Bisphosphonates
Bisphosphonates inhibit osteoclastic bone resorption by to hydroxyapatite sites, especially at sites undergoing active bone resorption. The exact mechanism for how bisphosphonates provide pain relieve in complex regional pain syndrome remains unclear, although there are a number of potential mechanisms that have been proposed and the true mechanism is likely multifactorial in nature given the limited degree of bone turnover noted in complex regional pain syndrome 222. Regardless, there appears to be a degree of evidence indicating the benefit of bisphosphonates in reducing pain scores in comparison to placebo; however, many of the early trials were often lacking in power. In addition, a lack of true understanding behind the mechanism of action for therapeutic benefit with bisphosphonates limits the ability to identify ideal drug and dosing regimens 223, 199. However, a randomized, double-blind, placebo-controlled study comparing four doses of intravenous (IV) neridronate to placebo found statistically significant decreases in Visual Analogue Scale (VAS) pain scores in the study of 82 patients. The authors posit that these results may be linked to bisphosphonate reduction of the increased bone turnover and subsequent marrow edema seen in complex regional pain syndrome that may contribute to chronic pain. Additionally, locally accumulated drug may interfere with the inflammatory pathways and subsequent pain generation by decreasing lactate concentration and acidosis. Interestingly, some patients also experienced permanent remission of their disease; however, it should be noted that these patients were identified early in their clinical course, an important caveat to the efficacy of bisphosphonates in treating complex regional pain syndrome. Regardless, the significant efficacy and limited adverse effects of the medications suggest an importance in incorporation of bisphosphonates as standard of care when initiating treatment for complex regional pain syndrome patients 222.
Ketamine
Ketamine is a strong anesthetic with an N-methyl-D-aspartate (NMDA) receptor antagonist has a notable role in the modulation of the wind-up response to noxious stimuli that results in central sensitization classically seen in chronic pain conditions. There is also evidence of a role in immunomodulation resulting in the down-regulation of neuroinflammatory markers; therefore, it remains unclear if the potential role of ketamine in complex regional pain syndrome treatment is relegated solely to the effects of NMDA antagonism resulting in down-regulation of central sensitization or if there are additional factors involved 224, 225. Investigators are using low doses of ketamine given intravenously for several days to either reduce substantially or eliminate the chronic pain of complex regional pain syndrome. In certain clinical settings, ketamine has been shown to be useful in treating pain that does not respond well to other treatments. Regardless, the current body of evidence is lacking in supporting the efficacy of ketamine in the treatment of complex regional pain syndrome with the majority of studies lacking in power or utilizing different modes of administration, making it difficult to determine the level of evidence supporting ketamine 226, 227, 224.
Botulinum Toxin A
The use of Botulinum Toxins A and B have been an emerging add-on (adjunct) to local anesthetics for sympathectomies and treatment of dystonia or spasticity in the treatment of complex regional pain syndrome, with most studies published starting in 2008. The mechanism of this therapy was hypothesized to be slightly different depending on the use for treating dystonia and spasticity or for a sympathectomy. In dystonia, the role of botulinum toxin in therapy is due to its function in disrupting the SNARE complex and inhibition of exocytosis. It prevents the release of acetylcholine and causes flaccid paralysis, effectively relieving the pain from muscle dystonia and spastic contractility 228, 229. A retrospective case review of the use of Botulinum Toxin A (Botox) in patients with neck or upper limb girdle muscles with dystonia and spasm showed that there was significant pain relief in 97% of the patients over the first 4 weeks after treatment. Given the efficacy of botulinum toxin in inhibition of cholinergic circuitry, several studies have expanded its use to blocking cholinergic signaling at the sympathetic ganglia in complex regional pain syndrome patients. The majority of studies have been small case-control or retrospective studies regarding the efficacy of Botulinum Toxin A (Botox) and Botulinum Toxin B (BTX-B), either by themselves or in conjunction with local anesthetic. While these studies range from retrospective studies to prospective randomized controlled trials, they all show moderate improvements in pain scores over the short term with either botulinum toxin alone or with local anesthetic. They also reliably show that the use of botulinum toxin has a greater duration of efficacy than the use of local anesthetic alone 230. A meta-analysis performed in 2020 which evaluated randomized controlled trials provided good evidence for the efficacy of Botulinum Toxin A (Botox) for pain relief across neuropathic and muscle based pain syndromes 231. There are numerous other studies that suggest the use of Botulinum Toxin A (Botox) and Botulinum Toxin B (BTX-B) depending on the location of the sympathectomy, but there is not yet strong evidence for the use of one or the other purely based on the site of complex regional pain syndrome being treated 232, 233. There is currently a study underway with very similar parameters looking at the impact to quality of life and disability scores in complex regional pain syndrome patients 234. While Botulinum Toxin A (Botox) has significant therapeutic potential, there are concerns about possible extravasation into surrounding ganglia which could lead to neurologic complications as well as discrepancies between industry and non-industry-sponsored studies 235.
Antioxidants
In the search for improved pharmacological treatments for complex regional pain syndrome, the use of antioxidants has been increasingly studied over the past ten years, spanning the fields of rheumatology, anesthesiology and orthopedics. This is primarily regarding complex regional pain syndrome Type 1, with an established injury or trauma, in many cases from surgery. Eisenberg et al. 236 in 2008 demonstrated that when comparing complex regional pain syndrome 1 and control patients, the patients with complex regional pain syndrome had significantly elevated salivary peroxidase, superoxide dismutase activity, uric acid and total antioxidant status compared to controls. In 2014 Baykal et al. 237 had similar findings, showing that superoxide dismutase, glutathione peroxidase and glutathione s-transferase activity were all significantly elevated in patients who developed complex regional pain syndrome Type 1 when compared to patients who did not. Several studies have been done regarding the prophylactic treatment of surgical patients with Vitamin C post-operatively. These have found that prophylaxis with 500 mg to 1 g of Vitamin C daily post operatively from total knee arthroplasty, foot and ankle surgery, and subacromial shoulder surgery had a statistically significant effect on the development of complex regional pain syndrome 1 238, 239, 240. Patients who received Vitamin C were less likely to develop complex regional pain syndrome, considered to be through the role of Vitamin C in stabilizing reactive oxygen species and in turn reducing inflammation. A meta-analysis of randomized controlled trials using vitamin C for complex regional pain syndrome prophylaxis found a similar reduction in the risk of complex regional pain syndrome development 241. In all, it is safe to say that Vitamin C perioperatively could be a valuable tool in the prevention of complex regional pain syndrome Type 1, especially in the elective surgical setting, ultimately reducing the incidence of this often-debilitating disease.
Sympathetic nerve block
Some individuals report temporary pain relief from sympathetic nerve blocks, but there is no published evidence of long-term benefit. Sympathetic blocks involve injecting an anesthetic next to the spine to directly block the activity of sympathetic nerves and improve blood flow.
It is thought that one of the underlying pathophysiologic mechanisms of complex regional pain syndrome is sympathetic hyperactivity. The most common sympathetic blocks for complex regional pain syndrome are the stellate ganglion sympathetic blocks used to treat upper extremity symptoms while lumbar sympathetic nerve blocks are used to treat lower extremity symptoms 148. Although the quality of the evidence was low, a 2013 Cochrane review found that sympathetic blocks combined with local anesthetic were ineffective at reducing complex regional pain syndrome-related pain 242. A more recent Cochrane review in 2016 was unable to make any firm judgments regarding the effectiveness of this type of treatment for complex regional pain syndrome 242. In 2019 a study looked at the relationship between sympathetic blocks and spinal cord stimulation and found that the effects of a sympathetic block did not predict the success of a spinal cord stimulator 243.
Surgical sympathectomy
A surgical sympathectomy is a procedure that cuts or clamps the sympathetic nerve chain to treat conditions such as excessive sweating, facial blushing, some chronic pain conditions, and Raynaud phenomenon. A surgeon cuts or clamps the sympathetic nerve chain, which runs along the spine in the chest and controls the body’s fight-or-flight response. Sympathectomy is a last resort when other treatments, such as medicines or antiperspirants, have not worked. The use of this operation that destroys some of the nerves is controversial. Some experts think it is unwarranted and makes complex regional pain syndrome worse, whereas others report a favorable outcome. Sympathectomy should be used only in individuals whose pain is dramatically relieved (although temporarily) by sympathetic nerve blocks.
Spinal cord stimulation
Placing stimulating electrodes through a needle into the epidural space to deliver electric stimulation to the spinal cord’s dorsal column provides a tingling sensation in the painful area. Electrodes may be placed temporarily for a few days in order to assess whether spinal cord stimulation (SCS) is likely to be helpful. Minor surgery is required to implant all the parts of the stimulator, battery, and electrodes under the skin on the torso. Some spinal cord stimulation (SCS) devices use an external pulse generator, but typically the electrodes are connected to an implanted pulse generator. Several mechanisms of action for spinal cord stimulation (SCS) have been proposed, including vasodilation, reversal of cortical maladaptive neuroplastic changes, adrenergic inhibition, and inhibition of nociceptive neural conduction in the spinal cord. A comprehensive outcome specific review of the use of spinal cord stimulation specifically for complex regional pain syndrome was published in 2017 244. High-level evidence continues to support spinal cord stimulation (SCS) beneficial and successful role in the treatment of complex regional pain syndrome patients’ perceived pain relief, pain score, and quality of life 244. Functional improvements were noted with less evidence for complex regional pain syndrome resolution, sleep hygiene improvement, positive psychological impact, and analgesic sparing effects 244. A 2021 study looking at the long-term outcome of spinal cord stimulation (SCS) in complex regional pain syndrome did not find statistical functional improvement or a reduction in opioid or neuropathic pain medication use 245. However, they did note that 70% of the patients continued to use their spinal cord stimulation (SCS) devices at the eight-year follow-up 245. Approximately 25 percent of individuals develop equipment problems that may require additional surgeries.
Dorsal root ganglion stimulation
The dorsal root ganglion serves as an ideal target given its role in processing and transmitting sensory information from the periphery to the central nervous system. In animal models of chronic pain, pathophysiologic changes in the dorsal root ganglion have been noted, including altered expression of various genes that may play a role in the hyperexcitability of neurons involved in the nociceptive pathway. An innovative neuromodulation technique for the treatment of chronic pain targets the dorsal root ganglion (DRG) rather than the spinal cord. The exact mechanism of how dorsal root ganglion stimulation exerts its beneficial effects remains unknown, although it may be related to the modulation of a variety of cells housed within the The dorsal root ganglion serves as an ideal target given its role in processing and transmitting sensory information from the periphery to the central nervous system. In animal models of chronic pain, pathophysiologic changes in the dorsal root ganglion have been noted, including altered expression of various genes that may play a role in the hyperexcitability of neurons involved in the nociceptive pathway 246, 247, 248, 249.
As opposed to conventional spinal cord stimulation (SCS), dorsal root ganglion stimulation enables a more targeted application of neurostimulation due to the dorsal root ganglion’s more peripheral location and ease of access. For the treatment of lower extremity pain in complex regional pain syndrome, dorsal root ganglion stimulation received approval from the US Food and Drug Administration (FDA) in 2016. According to a recent pooled analysis study, dorsal root ganglion stimulation was safe and effective for complex regional pain syndrome, resulting in a 4.9-point mean decrease in complex regional pain syndrome type 1 pain intensity 250. In 2017 the ACCURATE study compared spinal cord stimulation (SCS) and dorsal root ganglion stimulation in 152 patients with complex regional pain syndrome 248. Dorsal root ganglion stimulation was found to be more efficient than conventional spinal cord stimulation (SCS) in this multicenter randomized trial for reducing pain and enhancing quality of life in complex regional pain syndrome patients 248.
Other types of neural stimulation
Neurostimulation can be delivered at other locations along the pain pathway, not only at the spinal cord. These include near injured nerves (peripheral nerve stimulators), outside the membranes of the brain (motor cortex stimulation with dural electrodes), and within the parts of the brain that control pain (deep brain stimulation). A recent option involves the use of magnetic currents applied externally to the brain (known as repetitive Transcranial Magnetic Stimulation, or rTMS). A similar method that uses transcranial direct electrical stimulation is also being investigated. These stimulation methods have the advantage of being non-invasive, with the disadvantage that repeated treatment sessions are needed.
Epidural infusion
Epidural infusion is an alternative therapy to provide pain control, by allowing one to vary local anesthetic concentration and infusion dose to be titrated to the desired effect 131. Adjuvant (add-on) medications, such as clonidine with the addition of opioids, can be added to provide additional spinal analgesia and to potentiate the degree of relief 131. The most commonly used combination of epidural medications today includes clonidine with bupivacaine 131. Opioids can be added to the mixture if the pain relief is inadequate or if the local anesthetic concentration required to produce pain relief also prohibits ambulation or full participation in the physiotherapy program 131.
The primary benefit of continuous regional analgesia is that one is able to effectively titrate to the necessary degree of relief and promote active physical therapy as tolerated 131. Furthermore, with patient-activated bolus programming, these continuous regional techniques allow patients to self-administer small boluses for optimal analgesia as the pain levels fluctuate 131. Either before or after a strenuous exercise program, patients may experience elevations in pain, swelling, or allodynia. The ability for patients to readily self-administer extra doses of medication within certain pre-programmed parameters will improve patient satisfaction and optimize pain control 131. Rauck et al. 251 performed a randomized, blinded, placebo-controlled trial utilizing epidural clonidine. They randomized 26 patients with complex regional pain syndrome to receive daily epidural infusions (for 3 consecutive days) of clonidine 300 or 600 mcg, or placebo 251 If patients responded to the clonidine with analgesia and did not respond to placebo, they were placed on an open label infusion for a mean of 32 days at a mean dose of 32 mcg/hour 251. All patients had substantial relief with both the 300 and 700 microgram doses. Of the 26 patients, 19 elected to receive continuous infusions of clonidine for an average of 43 days with an average dose 32 ± 6 micrograms per hour 251. Seventeen of nineteen patients had statistically significant improvement in pain. Side effects were dizziness, dry mouth, mouth sores, and nausea. Six of 19 patients developed catheter related infection, and one developed meningitis 251.
Cooper et al. 252 studied 14 patients in a prospective open label trial and demonstrated improved pain relief and range of motion in patients receiving an epidural bupivacaine-opioid mixture for an average of 4 days. Thirteen of fourteen patients had significant improvement, with 11 of the 14 achieving “resolution of their complex regional pain syndrome” by the end of the trial with no activity restrictions 252. Konig et al. 253 studied 26 patients by using continuous cervical epidural analgesia of bupivacaine (0.25%) for seven days coupled with physical therapy. Eighty-three percent of patients had “improvement in pain”. Edema, sweating abnormalities, and dysfunction of the hand responded particularly well. Sixty-three percent of patients considered their condition to be acceptable whereas only 8% were completely pain free. Reduction in pain medications usage was also noted. Finally, Bucheit and Crews 254 described a single case report where continuous epidural infusion markedly improved range of motion.
The reported rates of infection in epidural catheters used to treat complex regional pain syndrome are as high as 31% 251. Therefore, epidural catheters meant for longer-term use should be performed as minor surgical procedures that require standard surgical sterility techniques. Catheters should be tunneled under the skin and away from the midline entrance point to the spine to minimize the colonization by bacteria that is inherently a greater risk with extended duration infusions. Standard catheter dressings, such as those required for extended central venous catheters, should be followed and dressings should be changed weekly. The hallmarks of an epidural abscess include the triad of back pain, sensorimotor loss, and loss of bowel and bladder function. Epidural abscesses may have earlier prodromal symptoms such as fever, neck pain, or photophobia 255. Careful attention to early symptoms is paramount for early diagnosis. A previous study has demonstrated a catheter related infection rate of 19 out of 350 patients. All of these patients were treated with antibiotics and catheter removal, and none required surgical intervention 255.
Intrathecal drug pumps
An intrathecal drug pump, also known as a pain pump, is a surgically implanted device that delivers painrelieving medications directly into the spinal fluid to treat chronic pain and spasticity. These medication may include opioids, local anesthetic agents, clonidine, and baclofen. The pump is about the size of a hockey puck and is implanted under the skin in the abdomen, flank, or axilla. A catheter is then inserted into the spinal cord and connected to the pump. Intrathecal pumps are often used to treat chronic pain caused by conditions such as cancer, neuropathic pain, failed back surgery, cerebral palsy, multiple sclerosis, stroke, brain injury, and spinal cord injury. Intrathecal pumps can provide more effective pain relief with fewer side effects than oral medication.
Intrathecal analgesia has been studied to a lesser extent when compared to epidural analgesia. Lundborg 256 reported a series of three patients with refractory complex regional pain syndrome, who did not have a favorable clinical response to intrathecal bupivacaine. In spite of initial analgesia, all patients demonstrated a progression of their complex regional pain syndrome 256. In a small subset of patients (n = 7) with refractory complex regional pain syndrome and severe dystonia, van Hilten et al. 257 demonstrated analgesia and functional restoration after a bolus of intrathecal baclofen injected in a double-blind fashion followed by intrathecal infusion. But van Hilten et al 257 stopped using intrathecal baclofen infusions due to what they deemed as unacceptable side effects. Intrathecal infusion for complex regional pain syndrome without dystonia has only limited supporting literature 257.
The major risk associated with these infusion techniques is the rate of infection, which remains to be defined by further prospective study on infusion techniques in complex regional pain syndrome patients.
Topical Treatments
Topical treatments must be distinguished from transdermal formulations such as the fentanyl or clonidine patches that deliver systemic medication through the skin. Topical medications remain local, reaching dermal nerve endings, blood vessels, and other cells in the skin. Topical medications are appealing by virtue of their relative lack of systemic effects; rashes and allergies are their only major adverse effect. Topical options to consider for complex regional pain syndrome include the 5% lidocaine impregnated patch, the capsaicin and dimethylsulfoxide (DMSO 131.
Some clinicians endorse the use Eutectic Mixture of Local Anesthetics (EMLA) for patients with complex regional pain syndrome 258, but it must be applied under an occlusive cover (e.g., plastic food wrap) to maximize penetration. The 5% lidocaine patch is FDA-approved for treating postherpetic neuralgia (PHN), and is available in generic formulation 259. It may have efficacy in some local or focal complex regional pain syndrome phenomena such as allodynia 260.
Capsaicin, the vanilloid compound in chili peppers, is a highly selective agonist for the Transient Receptor Potential channel, Vanilloid-receptor type 1 (TRPV1) that is expressed on central and peripheral terminals of nociceptive primary sensory neurons 261. Topical capsaicin causes activation followed by dying-back of nociceptive nerve endings by allowing unchecked cation influx 261. Capsaicin use is limited by the painful burning sensation it evokes at the site of application until the site becomes denervated. In a randomized controlled trial, topical capsaicin showed modest efficacy for post herpetic neuralgia 262. A preliminary study of high-dose topical capsaicin plus regional anesthesia for complex regional pain syndrome demonstrated partial efficacy 263. Some doctors have found topical capsaicin to be intolerably painful, somewhat messy, and unacceptable to most patients 264, 265, 266, 267, 268. In 2009, the FDA approved a high concentration 8% capsaicin patch for treating post herpetic neuralgia once every 3 months 269. Capsaicin is applied to the painful area for 1 hour after topical local anesthesia. Two additional well-powered randomized controlled trials were positive for high-versus low-dose capsaicin in peripheral neuropathic pain, including in HIV-associated distal sensory polyneuropathy 270, 271. However, in complex regional pain syndrome, exacerbated pain may render this treatment unsuitable 272.
Dimethyl sulfoxide (DMSO) is a free radical-scavenging agent. In a systematic review DMSO (50% cream for 2 months) provided significant complex regional pain syndrome symptom reduction when compared with placebo 273, however pain intensity was not improved 274.
Emerging treatments
Emerging treatments for complex regional pain syndrome include:
- Intravenous immunoglobulin (IVIG)
Researchers in United Kingdom report low-dose IVIG reduced pain intensity in a small trial of 13 patients with complex regional pain syndrome for 6 to 30 months who did not respond well to other treatments. Those who received intravenous immunoglobulin (IVIG) had a greater decrease in pain scores than those receiving saline during the following 14 days after infusion.
A randomized controlled trial of low-dose (0.5 g/kg) intravenous immunoglobulin (IVIG) treatment for persistent complex regional pain syndrome indicated efficacy 275. However, a subsequent larger trial was negative 276. Although there is some anecdotal evidence for the efficacy of high-dose IVIG this has not been formally tested.
- Plasma exchange therapy
Plasma exchange has been reported effective in several case series 277 and consequently complex regional pain syndrome has been added to the list of possible indications by the international plasma exchange society (ASFA) 278. Plasma exchange treatment, although providing pain improvement in some cases, requires use of long repeat-exchange cycles (e.g., eight exchange treatments over 4 weeks) and is unfortunately complicated regularly by pain increase at the venous access site.
- Graded Motor imagery
Several studies have demonstrated the benefits of graded motor imagery therapy for complex regional pain syndrome pain. Individuals do mental exercises including identifying left and right painful body parts while looking into a mirror and visualizing moving those painful body parts without actually moving them.
Several alternative therapies have been used to treat other painful conditions. Options include behavior modification, acupuncture, relaxation techniques (such as biofeedback, progressive muscle relaxation, and guided motion therapy), and chiropractic treatment.
Amputation
The role of amputation in chronic complex regional pain syndrome type 1 is a potentially viable one, specifically for those patients who suffer from intractable and refractory symptoms despite numerous multi-modal therapeutic interventions 128. Although not without their own risks as well as the potential for recurrence of complex regional pain syndrome in the remaining limb or other limbs, several studies have noted patient satisfaction with the benefits of amputation 128. These retrospective reviews are relatively limited in power; however, they suggest a potential role of amputation for those patients who have exhausted alternative options 279, 280, 281. There may be additional benefit in selecting potential amputation candidates for their resilience, as a study conducted by Bodde et al. 207 found that patients with higher resilience scores correlated with higher quality of life and lower psychological distress status post limb amputation. Regardless of the potential benefits, the decision to pursue amputation should be taken in the context of the patient overall, recognizing and acknowledging the associated complications of amputation as well as the risk for minimal relief to recurrence of complex regional pain syndrome 128.
Living with complex regional pain syndrome
With early treatment, you may keep complex regional pain syndrome from getting worse. Sometimes the condition improves. If treatment is started early enough, the symptoms may completely go away. People with long-lasting, more severe symptoms often don’t respond to treatment. These people may benefit from a pain management program aimed specifically at dealing with chronic pain.
Take care of your physical and mental health by following these suggestions:
- Maintain normal daily activities as best you can.
- Pace yourself and be sure to get the rest that you need.
- Stay connected with friends and family.
- Continue to pursue hobbies that you enjoy and are able to do.
If complex regional pain syndrome makes it difficult for you to do things you enjoy, ask your doctor about ways to get around the obstacles.
Keep in mind that your physical health can directly affect your mental health. Denial, anger and frustration are common with chronic illnesses.
At times, you may need more tools to deal with your emotions. A therapist, behavioral psychologist or other professional may be able to help you put things in perspective. They also may be able to teach you coping skills, such as relaxation or meditation techniques.
Sometimes joining a support group, where you can share experiences and feelings with other people, is a good approach. Ask your doctor what support groups are available in your community.
- Dydyk AM, Conermann T. Chronic Pain. [Updated 2024 May 6]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK553030[↩][↩][↩]
- Kemler MA, de Vet HC, Barendse GA, van den Wildenberg FA, van Kleef M. Effect of spinal cord stimulation for chronic complex regional pain syndrome Type I: five-year final follow-up of patients in a randomized controlled trial. J Neurosurg. 2008 Feb;108(2):292-8. doi: 10.3171/JNS/2008/108/2/0292[↩]
- Spinal cord stimulation. Medline Plus. https://medlineplus.gov/ency/article/007560.htm[↩][↩]
- Dydyk AM, Tadi P. Spinal Cord Stimulator Implant. [Updated 2023 Jul 3]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK555994[↩][↩][↩][↩]
- Deer TR, Mekhail N, Provenzano D, et al. Neuromodulation Appropriateness Consensus Committee. The appropriate use of neurostimulation: avoidance and treatment of complications of neurostimulation therapies for the treatment of chronic pain. Neuromodulation Appropriateness Consensus Committee. Neuromodulation. 2014 Aug;17(6):571-97; discussion 597-8. doi: 10.1111/ner.12206[↩]
- Buchser E, Durrer A, Albrecht E. Spinal cord stimulation for the management of refractory angina pectoris. J Pain Symptom Manage. 2006 Apr;31(4 Suppl):S36-42. doi: 10.1016/j.jpainsymman.2005.12.008[↩]
- Andréll P, Yu W, Gersbach P, Gillberg L, Pehrsson K, Hardy I, Ståhle A, Andersen C, Mannheimer C. Long-term effects of spinal cord stimulation on angina symptoms and quality of life in patients with refractory angina pectoris–results from the European Angina Registry Link Study (EARL). Heart. 2010 Jul;96(14):1132-6. doi: 10.1136/hrt.2009.177188[↩]
- Fedoroff IC, Blackwell E, Malysh L, McDonald WN, Boyd M. Spinal cord stimulation in pregnancy: a literature review. Neuromodulation. 2012 Nov-Dec;15(6):537-41; discussion 541. doi: 10.1111/j.1525-1403.2012.00448.x[↩]
- Hayek SM, Veizi E, Hanes M. Treatment-Limiting Complications of Percutaneous Spinal Cord Stimulator Implants: A Review of Eight Years of Experience From an Academic Center Database. Neuromodulation. 2015 Oct;18(7):603-8; discussion 608-9. doi: 10.1111/ner.12312[↩][↩]
- Deer TR, Mekhail N, Provenzano D, Pope J, Krames E, Thomson S, Raso L, Burton A, DeAndres J, Buchser E, Buvanendran A, Liem L, Kumar K, Rizvi S, Feler C, Abejon D, Anderson J, Eldabe S, Kim P, Leong M, Hayek S, McDowell G 2nd, Poree L, Brooks ES, McJunkin T, Lynch P, Kapural L, Foreman RD, Caraway D, Alo K, Narouze S, Levy RM, North R; Neuromodulation Appropriateness Consensus Committee. The appropriate use of neurostimulation: avoidance and treatment of complications of neurostimulation therapies for the treatment of chronic pain. Neuromodulation Appropriateness Consensus Committee. Neuromodulation. 2014 Aug;17(6):571-97; discussion 597-8. doi: 10.1111/ner.12206[↩]
- Kumar K, Buchser E, Linderoth B, Meglio M, Van Buyten JP. Avoiding complications from spinal cord stimulation: practical recommendations from an international panel of experts. Neuromodulation. 2007 Jan;10(1):24-33. doi: 10.1111/j.1525-1403.2007.00084.x[↩]
- Osborne MD, Ghazi SM, Palmer SC, Boone KM, Sletten CD, Nottmeier EW. Spinal cord stimulator–trial lead migration study. Pain Med. 2011 Feb;12(2):204-8. doi: 10.1111/j.1526-4637.2010.01019.x[↩]
- Vallejo R, Kramer J, Benyamin R. Neuromodulation of the cervical spinal cord in the treatment of chronic intractable neck and upper extremity pain: a case series and review of the literature. Pain Physician. 2007 Mar;10(2):305-11. https://www.painphysicianjournal.com/current/pdf?article=Nzk0&journal=32[↩]
- Wolter T, Kieselbach K. Cervical spinal cord stimulation: an analysis of 23 patients with long-term follow-up. Pain Physician. 2012 May-Jun;15(3):203-12. https://www.painphysicianjournal.com/current/pdf?article=MTY3Nw%3D%3D&journal=67[↩]
- North RB, Kidd DH, Petrucci L, Dorsi MJ. Spinal cord stimulation electrode design: a prospective, randomized, controlled trial comparing percutaneous with laminectomy electrodes: part II-clinical outcomes. Neurosurgery. 2005 Nov;57(5):990-6; discussion 990-6. doi: 10.1227/01.neu.0000180030.00167.b9[↩]
- Henderson JM, Schade CM, Sasaki J, Caraway DL, Oakley JC. Prevention of mechanical failures in implanted spinal cord stimulation systems. Neuromodulation. 2006 Jul;9(3):183-91. doi: 10.1111/j.1525-1403.2006.00059.x[↩]
- Bedder MD, Bedder HF. Spinal cord stimulation surgical technique for the nonsurgically trained. Neuromodulation. 2009 Apr;12 Suppl 1:1-19. doi: 10.1111/j.1525-1403.2009.00194.x[↩][↩]
- Rudiger J, Thomson S. Infection rate of spinal cord stimulators after a screening trial period. A 53-month third party follow-up. Neuromodulation. 2011 Mar-Apr;14(2):136-41; discussion 141. doi: 10.1111/j.1525-1403.2010.00317.x[↩]
- Cameron T. Safety and efficacy of spinal cord stimulation for the treatment of chronic pain: a 20-year literature review. J Neurosurg. 2004 Mar;100(3 Suppl Spine):254-67. doi: 10.3171/spi.2004.100.3.0254[↩]
- Turnbull DK, Shepherd DB. Post-dural puncture headache: pathogenesis, prevention and treatment. Br J Anaesth. 2003 Nov;91(5):718-29. doi: 10.1093/bja/aeg231[↩]
- Costigan SN, Sprigge JS. Dural puncture: the patients’ perspective. A patient survey of cases at a DGH maternity unit 1983-1993. Acta Anaesthesiol Scand. 1996 Jul;40(6):710-4. doi: 10.1111/j.1399-6576.1996.tb04515.x[↩]
- Petraglia FW 3rd, Farber SH, Gramer R, Verla T, Wang F, Thomas S, Parente B, Lad SP. The Incidence of Spinal Cord Injury in Implantation of Percutaneous and Paddle Electrodes for Spinal Cord Stimulation. Neuromodulation. 2016 Jan;19(1):85-90. doi: 10.1111/ner.12370[↩]
- Merskey H, Bogduk N. Classification of chronic pain. 2nd ed. Seattle: IASP Press, 1994. p. 1.[↩][↩][↩][↩]
- Treede RD, Rief W, Barke A, Aziz Q, Bennett MI, Benoliel R, Cohen M, Evers S, Finnerup NB, First MB, Giamberardino MA, Kaasa S, Kosek E, Lavand’homme P, Nicholas M, Perrot S, Scholz J, Schug S, Smith BH, Svensson P, Vlaeyen JWS, Wang SJ. A classification of chronic pain for ICD-11. Pain. 2015 Jun;156(6):1003-1007. doi: 10.1097/j.pain.0000000000000160 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4450869[↩][↩][↩]
- Bonica JJ. The management of pain. Philadelphia: Lea & Febiger, 1953.[↩][↩]
- Raffaeli W, Tenti M, Corraro A, Malafoglia V, Ilari S, Balzani E, Bonci A. Chronic Pain: What Does It Mean? A Review on the Use of the Term Chronic Pain in Clinical Practice. J Pain Res. 2021 Mar 29;14:827-835. doi: 10.2147/JPR.S303186[↩][↩][↩]
- Institute of Medicine (US) Committee on Advancing Pain Research, Care, and Education. Relieving Pain in America: A Blueprint for Transforming Prevention, Care, Education, and Research. Washington (DC): National Academies Press (US); 2011. Available from: https://www.ncbi.nlm.nih.gov/books/NBK91497[↩][↩]
- Voscopoulos C, Lema M. When does acute pain become chronic? Br J Anaesth. 2010 Dec;105 Suppl 1:i69-85. doi: 10.1093/bja/aeq323[↩][↩]
- Treede RD, Rief W, Barke A, Aziz Q, Bennett MI, Benoliel R, Cohen M, Evers S, Finnerup NB, First MB, Giamberardino MA, Kaasa S, Korwisi B, Kosek E, Lavand’homme P, Nicholas M, Perrot S, Scholz J, Schug S, Smith BH, Svensson P, Vlaeyen JWS, Wang SJ. Chronic pain as a symptom or a disease: the IASP Classification of Chronic Pain for the International Classification of Diseases (ICD-11). Pain. 2019 Jan;160(1):19-27. doi: 10.1097/j.pain.0000000000001384[↩][↩]
- Treede RD, Rief W, Barke A, et al. A classification of chronic pain for ICD-11. Pain. 2015;156(6):1003–1007. doi:10.1097/j.pain.0000000000000160 https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4450869[↩]
- Clifford JC. Successful management of chronic pain syndrome. Can Fam Physician. 1993 Mar;39:549-59. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2379763/pdf/canfamphys00109-0087.pdf[↩]
- What is chronic pain? https://www.iasp-pain.org/resources/topics/chronic-pain/[↩][↩][↩]
- InformedHealth.org [Internet]. Cologne, Germany: Institute for Quality and Efficiency in Health Care (IQWiG); 2006-. Fibromyalgia: Learn More – Understanding chronic pain. [Updated 2022 May 24]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK492976[↩][↩]
- Nijs J., George S.Z., Clauw D.J., Fernández-de-las-Peñas C., Kosek E., Ickmans K., Fernández-Carnero J., Polli A., Kapreli E., Huysmans E., et al. Central sensitisation in chronic pain conditions: Latest discoveries and their potential for precision medicine. Lancet Rheumatol. 2021;3:e383–e392. doi: 10.1016/S2665-9913(21)00032-1[↩]
- Kregel J., Meeus M., Malfliet A., Dolphens M., Danneels L., Nijs J., Cagnie B. Structural and functional brain abnormalities in chronic low back pain: A systematic review. Semin. Arthritis Rheum. 2015;45:229–237. doi: 10.1016/j.semarthrit.2015.05.002[↩]
- Albrecht D.S., Forsberg A., Sandstrom A., Bergan C., Kadetoff D., Protsenko E., Lampa J., Lee Y.C., Hoglund C.O., Catana C., et al. Brain glial activation in fibromyalgia—A multi-site positron emission tomography investigation. Brain Behav. Immun. 2019;75:72–83. doi: 10.1016/j.bbi.2018.09.018[↩]
- Ji R.R., Xu Z.Z., Gao Y.J. Emerging targets in neuroinflammation-driven chronic pain. Nat. Rev. Drug Discov. 2014;13:533–548. doi: 10.1038/nrd4334[↩]
- Knotkova H., Hamani C., Sivanesan E., Le Beuffe M.F.E., Moon J.Y., Cohen S.P., Huntoon M.A. Neuromodulation for chronic pain. Lancet. 2021;397:2111–2124. doi: 10.1016/S0140-6736(21)00794-7[↩]
- PDQ Supportive and Palliative Care Editorial Board. Cancer Pain (PDQ®): Health Professional Version. 2024 Jul 25. In: PDQ Cancer Information Summaries [Internet]. Bethesda (MD): National Cancer Institute (US); 2002-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK65949[↩]
- American Academy of Family Physicians. Chronic Pain. https://familydoctor.org/condition/chronic-pain[↩][↩][↩]
- Chronic Pain. Medline Plus. https://medlineplus.gov/chronicpain.html[↩]
- Gagnier JJ, Oltean H, van Tulder MW, Berman BM, Bombardier C, Robbins CB. Herbal Medicine for Low Back Pain: A Cochrane Review. Spine (Phila Pa 1976). 2016 Jan;41(2):116-33. doi: 10.1097/BRS.0000000000001310[↩][↩][↩][↩]
- Cameron M, Chrubasik S. Oral herbal therapies for treating osteoarthritis. Cochrane Database Syst Rev. 2014 May 22;2014(5):CD002947. doi: 10.1002/14651858.CD002947.pub2[↩]
- Singh JA, Noorbaloochi S, Macdonald R, Maxwell LJ. Chondroitin for osteoarthritis. Cochrane Database Syst Rev. 2015;2017(6). doi:10.1002/14651858.CD005614.pub2[↩][↩]
- Watkins LR, Milligan ED, Maier SF. Glial activation: a driving force for pathological pain. Trends Neurosci. 2001;24(8):450–455. doi: 10.1016/s0166-2236(00)01854-3[↩]
- Tracey I, Bushnell MC. How neuroimaging studies have challenged us to rethink: is chronic pain a disease? J Pain. 2009;10(11):1113–1120. doi: 10.1016/j.jpain.2009.09.001[↩]
- Grace PM, Hutchinson MR, Maier SF, Watkins LR. Pathological pain and the neuroimmune interface. Nat Rev Immunol. 2014;14(4):217–231. doi: 10.1038/nri3621[↩]
- Turk DC, Gatchel RJ, Eds. Psychological Approaches to Pain Management: A Practitioner’s Handbook. 3rd ed. New York: The Guilford Press; 2018.[↩]
- Niv D, Devor M. Chronic pain as a disease in its own right. Pain Pract. 2004 Sep;4(3):179-81. doi: 10.1111/j.1533-2500.2004.04301.x[↩]
- Smith BH, Macfarlane GJ, Torrance N. Epidemiology of chronic pain, from the laboratory to the bus stop: time to add understanding of biological mechanisms to the study of risk factors in population-based research? Pain. 2007 Jan;127(1-2):5-10. doi: 10.1016/j.pain.2006.11.001[↩][↩]
- van Hecke O, Torrance N, Smith BH. Chronic pain epidemiology and its clinical relevance. Br J Anaesth. 2013 Jul;111(1):13-8. doi: 10.1093/bja/aet123[↩]
- van Hecke O, Torrance N, Smith BH. Chronic pain epidemiology – where do lifestyle factors fit in? Br J Pain. 2013 Nov;7(4):209-17. doi: 10.1177/2049463713493264[↩]
- Aigner C.J., Cinciripini P.M., Anderson K.O., Baum G.P., Gritz E.R., Lam C.Y. The Association of Pain With Smoking and Quit Attempts in an Electronic Diary Study of Cancer Patients Trying to Quit. Nicotine Tob. Res. 2016;18:1449–1455. doi: 10.1093/ntr/ntv118[↩][↩]
- Ditre J.W., Heckman B.W., Zale E.L., Kosiba J.D., Maisto S.A. Acute analgesic effects of nicotine and tobacco in humans: A meta-analysis. Pain. 2016;157:1373–1381. doi: 10.1097/j.pain.0000000000000572[↩][↩]
- Landmark T., Romundstad P., Dale O., Borchgrevink P.C., Vatten L., Kaasa S. Chronic pain: One year prevalence and associated characteristics (the HUNT pain study) Scand. J. Pain. 2017;4:182–187. doi: 10.1016/j.sjpain.2013.07.022[↩][↩]
- Jakobsson U., Larsson C. Smoking and chronic pain among people aged 65 years and older. Pain Pract. 2014;14:237–244. doi: 10.1111/papr.12067[↩]
- Goesling J., Brummett C.M., Meraj T.S., Moser S.E., Hassett A.L., Ditre J.W. Associations Between Pain, Current Tobacco Smoking, Depression, and Fibromyalgia Status Among Treatment-Seeking Chronic Pain Patients. Pain Med. 2015;16:1433–1442. doi: 10.1111/pme.12747[↩]
- Fishbain D.A., Cole B., Lewis J.E., Gao J. Is smoking associated with alcohol-drug dependence in patients with pain and chronic pain patients? An evidence-based structured review. Pain Med. 2012;13:1212–1226. doi: 10.1111/j.1526-4637.2012.01446.x[↩]
- Chapman S.L., Wu L.T. Associations between cigarette smoking and pain among veterans. Epidemiol. Rev. 2015;37:86–102. doi: 10.1093/epirev/mxu008[↩]
- Sanford N.N., Sher D.J., Xu X., Ahn C., D’Amico A.V., Aizer A.A., Mahal B.A. Alcohol use among patients with cancer and survivors in the United States, 2000–2017. J. Natl. Compr. Cancer Netw. 2020;18:69–79. doi: 10.6004/jnccn.2019.7341[↩]
- Thompson T., Oram C., Correll C.U., Tsermentseli S., Stubbs B. Analgesic effects of alcohol: A systematic review and meta-analysis of controlled experimental studies in healthy participants. J. Pain. 2017;18:499–510. doi: 10.1016/j.jpain.2016.11.009[↩][↩]
- Zale E.L., Maisto S.A., Ditre J.W. Interrelations between pain and alcohol: An integrative review. Clin. Psychol. Rev. 2015;37:57–71. doi: 10.1016/j.cpr.2015.02.005[↩]
- Boissoneault J., Lewis B., Nixon S.J. Characterizing chronic pain and alcohol use trajectory among treatment-seeking alcoholics. Alcohol. 2019;75:47–54. doi: 10.1016/j.alcohol.2018.05.009[↩]
- Nieto S.J., Green R., Grodin E.N., Cahill C.M., Ray L.A. Pain catastrophizing predicts alcohol craving in heavy drinkers independent of pain intensity. Drug Alcohol. Depend. 2021;218:108368. doi: 10.1016/j.drugalcdep.2020.108368[↩]
- Alonso-Blanco C, Fernández-de-Las-Peñas C, Morales-Cabezas M, Zarco-Moreno P, Ge HY, Florez-García M. Multiple active myofascial trigger points reproduce the overall spontaneous pain pattern in women with fibromyalgia and are related to widespread mechanical hypersensitivity. Clin J Pain. 2011 Jun. 27(5):405-13.[↩]
- Gupta A, Mayer EA, Fling C, et al. Sex-based differences in brain alterations across chronic pain conditions. J Neurosci Res. 2017 Jan 2. 95 (1-2):604-616.[↩]
- Durosaro O, Davis MD, Hooten WM, et al. Intervention for erythromelalgia, a chronic pain syndrome: comprehensive pain rehabilitation center, Mayo Clinic. Arch Dermatol. 2008 Dec. 144(12):1578-83.[↩]
- Chronic Pain. https://www.iasp-pain.org/resources/topics/chronic-pain/[↩]
- Tang NK, Salkovskis PM, Hodges A, et al. Chronic pain syndrome associated with health anxiety: a qualitative thematic comparison between pain patients with high and low health anxiety. Br J Clin Psychol. 2009 Mar. 48:1-20.[↩]
- Agency for Healthcare Research and Quality. Treatment of Osteoarthritis of the Knee: An Update Review. Comparative Effectiveness Review no. 190. Rockville, MD: Agency for Healthcare Research and Quality; 2017. [↩]
- Qaseem A, Wilt TJ, McLean RM, et al. Noninvasive treatments for acute, subacute, and chronic low back pain: a clinical practice guideline from the American College of Physicians. Annals of Internal Medicine. 2017;166(7):514-530.[↩]
- Hochberg MC, Altman RD, April KT, et al. American College of Rheumatology 2012 recommendations for the use of nonpharmacologic and pharmacologic therapies in osteoarthritis of the hand, hip, and knee. Arthritis Care & Research. 2012;64(4):465-474.[↩]
- Ford AC, Moayyedi P, Chey WD, et al. American College of Gastroenterology monograph on management of irritable bowel syndrome. American Journal of Gastroenterology. 2018;113(Suppl 2):1-18.[↩]
- Fenske JN, Berland DW, Chandran SK, et al. Pain Management [Internet]. Ann Arbor (MI): Michigan Medicine University of Michigan; 2021 Jan. Available from: https://www.ncbi.nlm.nih.gov/books/NBK572296[↩][↩][↩]
- Macedo L.G., Smeets R.J., Maher C.G., Latimer J., McAuley J.H. Graded activity and graded exposure for persistent nonspecific low back pain: A systematic review. Phys. Ther. 2010;90:860–879. doi: 10.2522/ptj.20090303[↩]
- van Weering M., Vollenbroek-Hutten M.M., Kotte E.M., Hermens H.J. Daily physical activities of patients with chronic pain or fatigue versus asymptomatic controls. A systematic review. Clin. Rehabil. 2007;21:1007–1023. doi: 10.1177/0269215507078331[↩]
- Fransen M., McConnell S., Harmer A.R., Van der Esch M., Simic M., Bennell K.L. Exercise for osteoarthritis of the knee. Cochrane Database Syst. Rev. 2015;1:Cd004376. doi: 10.1002/14651858.CD004376.pub3[↩]
- McNair P.J., Simmonds M.A., Boocock M.G., Larmer P.J. Exercise therapy for the management of osteoarthritis of the hip joint: A systematic review. Arthritis Res. Ther. 2009;11:R98. doi: 10.1186/ar2743[↩]
- Lin C.W., McAuley J.H., Macedo L., Barnett D.C., Smeets R.J., Verbunt J.A. Relationship between physical activity and disability in low back pain: A systematic review and meta-analysis. Pain. 2011;152:607–613. doi: 10.1016/j.pain.2010.11.034[↩]
- Hasenbring M.I., Verbunt J.A. Fear-avoidance and endurance-related responses to pain: New models of behavior and their consequences for clinical practice. Clin. J. Pain. 2010;26:747–753. doi: 10.1097/AJP.0b013e3181e104f2[↩]
- Geneen L.J., Moore R.A., Clarke C., Martin D., Colvin L.A., Smith B.H. Physical activity and exercise for chronic pain in adults: An overview of Cochrane Reviews. Cochrane Database Syst. Rev. 2017;4:Cd011279. doi: 10.1002/14651858.CD011279.pub3[↩][↩]
- Osteoarthritis in over 16s: diagnosis and management. https://www.nice.org.uk/guidance/ng226[↩]
- Lee H., Wiggers J., Kamper S.J., Williams A., O’Brien K.M., Hodder R.K., Wolfenden L., Yoong S.L., Campbell E., Haskins R., et al. Mechanism evaluation of a lifestyle intervention for patients with musculoskeletal pain who are overweight or obese: Protocol for a causal mediation analysis. BMJ Open. 2017;7:e014652. doi: 10.1136/bmjopen-2016-014652[↩]
- Vaegter H.B., Stoten M., Silseth S.L., Erlangsen A., Handberg G., Sondergaard S., Stenager E. Cause-specific mortality of patients with severe chronic pain referred to a multidisciplinary pain clinic: A cohort register-linkage study. Scand. J. Pain. 2019;19:93–99. doi: 10.1515/sjpain-2018-0094[↩]
- Macfarlane G.J., Barnish M.S., Jones G.T. Persons with chronic widespread pain experience excess mortality: Longitudinal results from UK Biobank and meta-analysis. Ann. Rheum. Dis. 2017;76:1815–1822. doi: 10.1136/annrheumdis-2017-211476[↩]
- Brain K., Burrows T.L., Rollo M.E., Chai L.K., Clarke E.D., Hayes C., Hodson F.J., Collins C.E. A systematic review and meta-analysis of nutrition interventions for chronic noncancer pain. J. Hum. Nutr. Diet. Off. J. Br. Diet. Assoc. 2018;32:198–225. doi: 10.1111/jhn.12601[↩][↩][↩][↩]
- Elma Ö., Tümkaya Yılmaz S., Nijs J., Clarys P., Coppieters I., Mertens E., Deliens T., Malfliet A. Proinflammatory Dietary Intake Relates to Pain Sensitivity in Chronic Nonspecific Low Back Pain: A Case-Control Study. J. Pain. 2024;25:350–361. doi: 10.1016/j.jpain.2023.08.015[↩]
- Elma Ö., Yilmaz S.T., Deliens T., Coppieters I., Clarys P., Nijs J., Malfliet A. Do Nutritional Factors Interact with Chronic Musculoskeletal Pain? A Systematic Review. J. Clin. Med. 2020;9:702. doi: 10.3390/jcm9030702[↩]
- Brain K., Burrows T.L., Bruggink L., Malfliet A., Hayes C., Hodson F.J., Collins C.E. Diet and Chronic Non-Cancer Pain: The State of the Art and Future Directions. J. Clin. Med. 2021;10:5203. doi: 10.3390/jcm10215203[↩][↩]
- Zhang T.T., Liu Z., Liu Y.L., Zhao J.J., Liu D.W., Tian Q.B. Obesity as a Risk Factor for Low Back Pain: A Meta-Analysis. Clin. Spine Surg. 2016;31:22–27. doi: 10.1097/BSD.0000000000000468[↩][↩]
- Hussain S.M., Urquhart D.M., Wang Y., Shaw J.E., Magliano D.J., Wluka A.E., Cicuttini F.M. Fat mass and fat distribution are associated with low back pain intensity and disability: Results from a cohort study. Arthritis Res. Ther. 2017;19:26. doi: 10.1186/s13075-017-1242-z[↩][↩][↩]
- Williams A., Wiggers J., O’Brien K.M., Wolfenden L., Yoong S., Campbell E., Robson E., McAuley J., Haskins R., Kamper S.J., et al. A randomised controlled trial of a lifestyle behavioural intervention for patients with low back pain, who are overweight or obese: Study protocol. BMC Musculoskelet. Disord. 2016;17:70. doi: 10.1186/s12891-016-0922-1[↩][↩]
- Reme S.E., Tveito T.H., Harris A., Lie S.A., Grasdal A., Indahl A., Brox J.I., Tangen T., Hagen E.M., Gismervik S., et al. Cognitive Interventions and Nutritional Supplements (The CINS Trial): A Randomized Controlled, Multicenter Trial Comparing a Brief Intervention With Additional Cognitive Behavioral Therapy, Seal Oil, and Soy Oil for Sick-Listed Low Back Pain Patients. Spine. 2016;41:1557–1564. doi: 10.1097/BRS.0000000000001596[↩]
- Roffey D.M., Ashdown L.C., Dornan H.D., Creech M.J., Dagenais S., Dent R.M., Wai E.K. Pilot evaluation of a multidisciplinary, medically supervised, nonsurgical weight loss program on the severity of low back pain in obese adults. Spine J. Off. J. N. Am. Spine Soc. 2011;11:197–204. doi: 10.1016/j.spinee.2011.01.031[↩][↩]
- Shiri R., Karppinen J., Leino-Arjas P., Solovieva S., Viikari-Juntura E. The association between obesity and low back pain: A meta-analysis. Am. J. Epidemiol. 2010;171:135–154. doi: 10.1093/aje/kwp356[↩][↩][↩]
- Vismara L., Menegoni F., Zaina F., Galli M., Negrini S., Capodaglio P. Effect of obesity and low back pain on spinal mobility: A cross sectional study in women. J. Neuroeng. Rehabil. 2010;7:3. doi: 10.1186/1743-0003-7-3[↩]
- Hershkovich O., Friedlander A., Gordon B., Arzi H., Derazne E., Tzur D., Shamis A., Afek A. Associations of body mass index and body height with low back pain in 829,791 adolescents. Am. J. Epidemiol. 2013;178:603–609. doi: 10.1093/aje/kwt019[↩]
- Paulis W.D., Silva S., Koes B.W., van Middelkoop M. Overweight and obesity are associated with musculoskeletal complaints as early as childhood: A systematic review. Obes. Rev. An. Off. J. Int. Assoc. Study Obes. 2014;15:52–67. doi: 10.1111/obr.12067[↩]
- Dario A.B., Ferreira M.L., Refshauge K., Sanchez-Romera J.F., Luque-Suarez A., Hopper J.L., Ordonana J.R., Ferreira P.H. Are obesity and body fat distribution associated with low back pain in women? A population-based study of 1128 Spanish twins. Eur. Spine J. 2016;25:1188–1195. doi: 10.1007/s00586-015-4055-2[↩]
- Hall M., Castelein B., Wittoek R., Calders P., Van Ginckel A. Diet-induced weight loss alone or combined with exercise in overweight or obese people with knee osteoarthritis: A systematic review and meta-analysis. Semin. Arthritis Rheum. 2018;48:765–777. doi: 10.1016/j.semarthrit.2018.06.005[↩]
- Messier S.P., Beavers D.P., Mihalko S.L., Miller G.D., Lyles M.F., Hunter D.J., Carr J.J., Eckstein F., Guermazi A., Loeser R.F., et al. The effects of intensive dietary weight loss and exercise on gait in overweight and obese adults with knee osteoarthritis. The Intensive Diet and Exercise for Arthritis (IDEA) trial. J. Biomech. 2020;98:109477. doi: 10.1016/j.jbiomech.2019.109477[↩][↩][↩]
- Losina E., Smith K.C., Paltiel A.D., Collins J.E., Suter L.G., Hunter D.J., Katz J.N., Messier S.P. Cost-Effectiveness of Diet and Exercise for Overweight and Obese Knee Osteoarthritis Patients. Arthritis Care Res. 2018;71:855–864. doi: 10.1002/acr.23716[↩]
- Burgess E., Hassmen P., Welvaert M., Pumpa K.L. Behavioural treatment strategies improve adherence to lifestyle intervention programmes in adults with obesity: A systematic review and meta-analysis. Clin. Obes. 2017;7:105–114. doi: 10.1111/cob.12180[↩]
- Donnelly J.E., Blair S.N., Jakicic J.M., Manore M.M., Rankin J.W., Smith B.K. American College of Sports Medicine Position Stand. Appropriate physical activity intervention strategies for weight loss and prevention of weight regain for adults. Med. Sci. Sports Exerc. 2009;41:459–471. doi: 10.1249/MSS.0b013e3181949333[↩]
- Buesing S., Costa M., Schilling J.M., Moeller-Bertram T. Vitamin B12 as a Treatment for Pain. Pain Phys. 2019;22:e45–e52. doi: 10.36076/ppj/2019.22.E45[↩]
- Shipton E.A., Shipton E.E. Vitamin D and Pain: Vitamin D and Its Role in the Aetiology and Maintenance of Chronic Pain States and Associated Comorbidities. Pain Res. Treat. 2015;2015:904967. doi: 10.1155/2015/904967[↩]
- Lin Y-C, Wan L, Jamison RN. Using integrative medicine in pain management: an evaluation of current evidence. Anesthesia and Analgesia. 2017;125(6):2081-2093.[↩]
- Vickers AJ, Vertosick EA, Lewith G, et al. Acupuncture for chronic pain: update of an individual patient data meta-analysis. Journal of Pain. 2018;19(5):455-474.[↩]
- Nahin RL, Boineau R, Khalsa PS, Stussman BJ, Weber WJ. Evidence-based evaluation of complementary health approaches for pain management in the United States. Mayo Clinic Proceedings. 2016;91(9):1291-1306.[↩]
- Garza-Villareal E, Pando V, Vuust P, et al. Music-induced analgesia in chronic pain conditions: a systematic review and meta-analysis. Pain Physician. 2017;20(7):597-610.[↩]
- Field T. Massage therapy research review. Complement Ther Clin Pract. 2016 Aug;24:19-31. doi: 10.1016/j.ctcp.2016.04.005[↩]
- Nelson NL, Churilla JR. Massage Therapy for Pain and Function in Patients With Arthritis: A Systematic Review of Randomized Controlled Trials. Am J Phys Med Rehabil. 2017 Sep;96(9):665-672. doi: 10.1097/PHM.0000000000000712[↩]
- DXC. Braddom’s Physical Medicine and Rehabilitation 5th Edition. 5th ed. Elsevier; 2015.[↩]
- Cameron M, Chrubasik S. Oral herbal therapies for treating osteoarthritis. Cochrane Database Syst Rev. 2014;2014(5). doi:10.1002/14651858.CD002947.pub2[↩]
- Nugent SM, Morasco BJ, O’Neil ME, Freeman M, Low A, Kondo K, Elven C, Zakher B, Motu’apuaka M, Paynter R, Kansagara D. The Effects of Cannabis Among Adults With Chronic Pain and an Overview of General Harms: A Systematic Review. Ann Intern Med. 2017 Sep 5;167(5):319-331. doi: 10.7326/M17-0155[↩][↩]
- Daily JW, Yang M, Park S. Efficacy of Turmeric Extracts and Curcumin for Alleviating the Symptoms of Joint Arthritis: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. J Med Food. 2016 Aug;19(8):717-29. doi: 10.1089/jmf.2016.3705[↩]
- Chou R, Deyo R, Friedly J, Skelly A, Weimer M, Fu R, Dana T, Kraegel P, Griffin J, Grusing S. Systemic Pharmacologic Therapies for Low Back Pain: A Systematic Review for an American College of Physicians Clinical Practice Guideline. Ann Intern Med. 2017 Apr 4;166(7):480-492. doi: 10.7326/M16-2458[↩]
- Enthoven WT, Roelofs PD, Deyo RA, van Tulder MW, Koes BW. Non-steroidal anti-inflammatory drugs for chronic low back pain. Cochrane Database Syst Rev. 2016 Feb 10;2(2):CD012087. doi: 10.1002/14651858.CD012087[↩]
- Argoff CE. Topical analgesics in the management of acute and chronic pain. Mayo Clin Proc. 2013 Feb;88(2):195-205. doi: 10.1016/j.mayocp.2012.11.015[↩]
- Dowell D, Haegerich TM, Chou R. CDC Guideline for Prescribing Opioids for Chronic Pain–United States, 2016. JAMA. 2016 Apr 19;315(15):1624-45. doi: 10.1001/jama.2016.1464[↩]
- Watkins PB, Kaplowitz N, Slattery JT, Colonese CR, Colucci SV, Stewart PW, Harris SC. Aminotransferase elevations in healthy adults receiving 4 grams of acetaminophen daily: a randomized controlled trial. JAMA. 2006 Jul 5;296(1):87-93. doi: 10.1001/jama.296.1.87[↩]
- Holubek WJ, Kalman S, Hoffman RS. Acetaminophen-induced acute liver failure: results of a United States multicenter, prospective study. Hepatology. 2006 Apr;43(4):880; author reply 882. doi: 10.1002/hep.21106[↩]
- Jalan R, Williams R, Bernuau J. Paracetamol: are therapeutic doses entirely safe? Lancet. 2006 Dec 23;368(9554):2195-6. doi: 10.1016/S0140-6736(06)69874-7[↩]
- Cavalcante AN, Sprung J, Schroeder DR, Weingarten TN. Multimodal Analgesic Therapy With Gabapentin and Its Association With Postoperative Respiratory Depression. Anesth Analg. 2017 Jul;125(1):141-146. doi: 10.1213/ANE.0000000000001719[↩]
- Gomes T, Greaves S, van den Brink W, Antoniou T, Mamdani MM, Paterson JM, Martins D, Juurlink DN. Pregabalin and the Risk for Opioid-Related Death: A Nested Case-Control Study. Ann Intern Med. 2018 Nov 20;169(10):732-734. doi: 10.7326/M18-1136[↩]
- Lee M, Silverman SM, Hansen H, Patel VB, Manchikanti L. A comprehensive review of opioid-induced hyperalgesia. Pain Physician. 2011 Mar-Apr;14(2):145-61. https://www.painphysicianjournal.com/current/pdf?article=MTQ0Ng%3D%3D&journal=60[↩]
- Managing your chronic back pain. Medline Plus. https://medlineplus.gov/ency/patientinstructions/000516.htm[↩]
- Abd-Elsayed A, Stark CW, Topoluk N, Isaamullah M, Uzodinma P, Viswanath O, Gyorfi MJ, Fattouh O, Schlidt KC, Dyara O. A brief review of complex regional pain syndrome and current management. Ann Med. 2024 Dec;56(1):2334398. doi: 10.1080/07853890.2024.2334398[↩][↩][↩][↩][↩][↩][↩]
- Mangnus TJP, Bharwani KD, Dirckx M, Huygen FJPM. From a Symptom-Based to a Mechanism-Based Pharmacotherapeutic Treatment in Complex Regional Pain Syndrome. Drugs. 2022 Apr;82(5):511-531. doi: 10.1007/s40265-022-01685-4[↩]
- MG30.04 Complex regional pain syndrome. https://icd.who.int/browse/2024-01/mms/en#1834504950[↩]
- Harden RN, McCabe CS, Goebel A, Massey M, Suvar T, Grieve S, Bruehl S. Complex Regional Pain Syndrome: Practical Diagnostic and Treatment Guidelines, 5th Edition. Pain Med. 2022 Jun 10;23(Suppl 1):S1-S53. doi: 10.1093/pm/pnac046[↩][↩][↩][↩][↩][↩][↩][↩][↩][↩][↩]
- Veldman PH, Reynen HM, Arntz IE, Goris RJ. Signs and symptoms of reflex sympathetic dystrophy: prospective study of 829 patients. Lancet. 1993;342(8878):1012–1016. doi: 10.1016/0140-6736(93)92877-v[↩]
- Finnerup NB, Haroutounian S, Kamerman P, Baron R, Bennett DLH, Bouhassira D, Cruccu G, Freeman R, Hansson P, Nurmikko T, Raja SN, Rice ASC, Serra J, Smith BH, Treede RD, Jensen TS. Neuropathic pain: an updated grading system for research and clinical practice. Pain. 2016 Aug;157(8):1599-1606. doi: 10.1097/j.pain.0000000000000492[↩]
- Marinus J, Moseley GL, Birklein F, Baron R, Maihöfner C, Kingery WS, et al. Clinical features and pathophysiology of complex regional pain syndrome. Lancet Neurology 2011;10(7):637-48. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5511749/[↩]
- Stanton-Hicks M, Baron R, Boas R, Gordh T, Harden N, Hendler N, Koltzenburg M, Raj P, Wilder R. Complex Regional Pain Syndromes: guidelines for therapy. Clin J Pain. 1998 Jun;14(2):155-66. doi: 10.1097/00002508-199806000-00012[↩]
- Perez RS, Zollinger PE, Dijkstra PU, Thomassen-Hilgersom IL, Zuurmond WW, Rosenbrand KC, Geertzen JH; CRPS I task force. Evidence based guidelines for complex regional pain syndrome type 1. BMC Neurol. 2010 Mar 31;10:20. doi: 10.1186/1471-2377-10-20[↩]
- Merskey H, Bogduk N. Classification of Chronic Pain. 2nd (Revised) ed. IASP Task Force on Taxonomy, IASP Press, 2011; 4–7[↩]
- Taylor SS, Noor N, Urits I, Paladini A, Sadhu MS, Gibb C, Carlson T, Myrcik D, Varrassi G, Viswanath O. Complex Regional Pain Syndrome: A Comprehensive Review. Pain Ther. 2021 Dec;10(2):875-892. doi: 10.1007/s40122-021-00279-4. Epub 2021 Jun 24. Erratum in: Pain Ther. 2021 Dec;10(2):893-894. doi: 10.1007/s40122-021-00291-8[↩]
- Bruehl S. Complex regional pain syndrome. BMJ. 2015 Jul 29;351:h2730. https://www.bmj.com/content/351/bmj.h2730.long[↩]
- Giannotti S, Bottai V, Dell’Osso G, Bugelli G, Celli F, Cazzella N, Guido G. Algodystrophy: complex regional pain syndrome and incomplete forms. Clin Cases Miner Bone Metab. 2016 Jan-Apr;13(1):11-4. doi: 10.11138/ccmbm/2016.13.1.011[↩]
- Russo M, Georgius P, Santarelli DM. A new hypothesis for the pathophysiology of complex regional pain syndrome. Med Hypotheses. 2018 Oct;119:41-53. doi: 10.1016/j.mehy.2018.07.026[↩]
- Lloyd ECO, Dempsey B, Romero L. Complex Regional Pain Syndrome. Am Fam Physician. 2021 Jul 1;104(1):49-55. https://www.aafp.org/pubs/afp/issues/2021/0700/p49.html[↩]
- Kessler A, Yoo M, Calisoff R. Complex regional pain syndrome: An updated comprehensive review. NeuroRehabilitation. 2020;47(3):253-264. doi: 10.3233/NRE-208001[↩]
- de Mos M, de Bruijn AG, Huygen FJ, Dieleman JP, Stricker BH, Sturkenboom MC. The incidence of complex regional pain syndrome: a population-based study. Pain. 2007 May;129(1-2):12-20. doi: 10.1016/j.pain.2006.09.008[↩][↩][↩]
- Lewis JS, Kashif M, Maan A, Ciampi de Andrade D, Casey M, Moon JY, Lin CP, Danielsson L, Quek T, Díez Tafur R, Aloweidi A, Birklein F, Knudsen L, Goebel A. Global series: Complex regional pain syndrome: abstracts from the International Association for the Study of Pain complex regional pain syndrome SIG virtual symposia 2021. Pain Rep. 2023 Jan 16;8(1):e1056. doi: 10.1097/PR9.0000000000001056[↩][↩]
- Bruehl S, Chung OY. How common is complex regional pain syndrome-Type I? Pain. 2007 May;129(1-2):1-2. doi: 10.1016/j.pain.2007.02.017[↩]
- Beerthuizen A, Stronks DL, Van’t Spijker A, Yaksh A, Hanraets BM, Klein J, Huygen FJPM. Demographic and medical parameters in the development of complex regional pain syndrome type 1 (CRPS1): prospective study on 596 patients with a fracture. Pain. 2012 Jun;153(6):1187-1192. doi: 10.1016/j.pain.2012.01.026[↩]
- Misidou C, Papagoras C. Complex Regional Pain Syndrome: An update. Mediterr J Rheumatol. 2019 Mar 28;30(1):16-25. doi: 10.31138/mjr.30.1.16[↩][↩][↩][↩]
- Goebel A, Birklein F, Brunner F, et al. The valencia consensus-based adaptation of the IASP complex regional pain syndrome diagnostic criteria. Pain. 2021;162:2346–2348. doi: 10.1097/j.pain.0000000000002245[↩]
- Bruehl S. Complex regional pain syndrome. BMJ. 2015;351 doi: 10.1136/bmj.h2730. h2730[↩]
- Goebel A, Barker C, Birklein F, et al. Standards for the diagnosis and management of complex regional pain syndrome: results of a European Pain Federation task force. Eur J Pain. 2019;23:641–651. doi: 10.1002/ejp.1362[↩]
- Harden RN, McCabe CS, Goebel A, et al. Complex regional pain syndrome: practical diagnostic and treatment guidelines, 5th edition. Pain Med. 2022;23:1–53. doi: 10.1093/pm/pnac046[↩]
- Complex regional pain syndrome in adults (2nd edition). UK guidelines for diagnosis, referral and management in primary and secondary care 2018. https://www.rcp.ac.uk/media/4uijppdz/complex-regional-pain-syndrome-in-adults-second-edition_0.pdf[↩]
- Stanton-Hicks MD, Burton AW, Bruehl SP, Carr DB, Harden RN, Hassenbusch SJ, et al. An updated interdisciplinary clinical pathway for CRPS: report of an expert panel. Pain Practice 2002;2(1):1-16. https://www.ncbi.nlm.nih.gov/pubmed/17134466[↩]
- Bruehl S. An update on the pathophysiology of complex regional pain syndrome. Anesthesiology 2010;113(3):713-25. https://www.ncbi.nlm.nih.gov/pubmed/20693883[↩]
- Lewis JS, Kersten P, Mccabe CS, McPherson KM, Blake DR. Body perception disturbance: a contribution to pain in complex regional pain syndrome (CRPS). Pain 2007;133(1-3):111-9. https://www.ncbi.nlm.nih.gov/pubmed/17509761[↩]
- Ott S, Maihöfner C. Signs and Symptoms in 1,043 Patients with Complex Regional Pain Syndrome. J Pain. 2018 Jun;19(6):599-611. doi: 10.1016/j.jpain.2018.01.004[↩][↩]
- Fukushima FB, Bezerra DM, Villas Boas PJ, Valle AP, Vidal EI. Complex regional pain syndrome. BMJ. 2014 Jun 25;348:g3683. doi: 10.1136/bmj.g3683[↩]
- Harden RN, Oaklander AL, Burton AW, Perez RS, Richardson K, Swan M, Barthel J, Costa B, Graciosa JR, Bruehl S; Reflex Sympathetic Dystrophy Syndrome Association. Complex regional pain syndrome: practical diagnostic and treatment guidelines, 4th edition. Pain Med. 2013 Feb;14(2):180-229. doi: 10.1111/pme.12033[↩][↩][↩][↩]
- Wasner G, Schattschneider J, Heckmann K, Maier C, Baron R. Vascular abnormalities in reflex sympathetic dystrophy (CRPS I): mechanisms and diagnostic value. Brain. 2001 Mar;124(Pt 3):587-99. doi: 10.1093/brain/124.3.587[↩]
- Harden RN, Bruehl S, Stanton-Hicks M, Wilson PR. Proposed new diagnostic criteria for complex regional pain syndrome. Pain Med. 2007 May-Jun;8(4):326-31. doi: 10.1111/j.1526-4637.2006.00169.x[↩][↩]
- Williams K, Guarino A, Raja SN.. Chapter 27 – Complex regional pain syndrome. In: Benzon HT, Raja SN, Liu SS, et al., eds. Essentials of pain medicine. 4th ed. Philadelphia: Elsevier; 2018:223.e2–232.e2. doi: 10.1016/B978-0-323-40196-8.00027-9[↩][↩][↩][↩][↩][↩]
- Harden NR, Bruehl S, Perez RSGM, Birklein F, Marinus J, Maihofner C, Lubenow T, Buvanendran A, Mackey S, Graciosa J, Mogilevski M, Ramsden C, Chont M, Vatine JJ. Validation of proposed diagnostic criteria (the “Budapest Criteria”) for Complex Regional Pain Syndrome. Pain. 2010 Aug;150(2):268-274. doi: 10.1016/j.pain.2010.04.030[↩]
- Melf-Marzi A, Böhringer B, Wiehle M, Hausteiner-Wiehle C. Modern Principles of Diagnosis and Treatment in Complex Regional Pain Syndrome. Dtsch Arztebl Int. 2022 Dec 27;119(51-52):879-886. doi: 10.3238/arztebl.m2022.0358[↩][↩]
- Harden RN, Bruehl S, Perez RSGM, et al. Validation of proposed diagnostic criteria (the “Budapest Criteria”) for Complex Regional Pain Syndrome. Pain. 2010;150:268–74. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2914601/[↩]
- Devarajan J, Mena S, Cheng J. Mechanisms of complex regional pain syndrome. Front Pain Res (Lausanne). 2024 May 17;5:1385889. doi: 10.3389/fpain.2024.1385889[↩]
- Shim H, et al. Complex regional pain syndrome: a narrative review for the practising clinician. Br J Anaesth. 2019;123(2):e424–e433. doi: 10.1016/j.bja.2019.03.030[↩]
- Li TS, Wang R, Su X, Wang XQ. Effect and mechanisms of exercise for complex regional pain syndrome. Front Mol Neurosci. 2023 May 3;16:1167166. doi: 10.3389/fnmol.2023.1167166[↩]
- Ott S, Maihöfner C. Signs and symptoms in 1,043 patients with complex regional pain syndrome. J Pain. 2018;19(6):599–611. doi: 10.1016/j.jpain.2018.01.004[↩][↩][↩][↩]
- Mertz K, Trunzter J, Wu E, Barnes J, Eppler SL, Kamal RN. National trends in the diagnosis of CRPS after open and endoscopic carpal tunnel release. J Wrist Surg. 2019;08(03):209–214. doi: 10.1055/s-0039-1678674[↩]
- Jo Y-H, Kim K, Lee B-G, Kim J-H, Lee C-H, Lee K-H. Incidence of and risk factors for complex regional pain syndrome type 1 after surgery for distal radius fractures: a population-based study. Sci Rep. 2019 doi: 10.1038/S41598-019-41152-X[↩][↩][↩][↩]
- Bruehl S. An update on the pathophysiology of complex regional pain syndrome. Anesthesiology. 2010 Sep;113(3):713-25. doi: 10.1097/ALN.0b013e3181e3db38[↩]
- de Rooij AM, de Mos M, Sturkenboom MC, Marinus J, van den Maagdenberg AM, van Hilten JJ. Familial occurrence of complex regional pain syndrome. Eur J Pain. 2009 Feb;13(2):171-7. doi: 10.1016/j.ejpain.2008.04.004[↩]
- Shaikh SS, Goebel A, Lee MC, Nahorski MS, Shenker N, Pamela Y, Drissi I, Brown C, Ison G, Shaikh MF, Kuttikat A, Woods WA, Dixit A, Stouffer K, Clarke MC, Menon DK, Woods CG. Evidence of a genetic background predisposing to complex regional pain syndrome type 1. J Med Genet. 2024 Jan 19;61(2):163-170. doi: 10.1136/jmg-2023-109236[↩]
- Modarresi S, Aref-Eshghi E, Walton DM, MacDermid JC. Does a familial subtype of complex regional pain syndrome exist? Results of a systematic review. Can J Pain. 2019 Aug 1;3(1):157-166. doi: 10.1080/24740527.2019.1637249[↩]
- Jin EH, Zhang E, Ko Y, Sim WS, Moon DE, Yoon KJ, Hong JH, Lee WH. Genome-wide expression profiling of complex regional pain syndrome. PLoS One. 2013 Nov 14;8(11):e79435. doi: 10.1371/journal.pone.0079435[↩]
- Pohóczky K, Kun J, Szentes N, Aczél T, Urbán P, Gyenesei A, Bölcskei K, Szőke É, Sensi S, Dénes Á, Goebel A, Tékus V, Helyes Z. Discovery of novel targets in a complex regional pain syndrome mouse model by transcriptomics: TNF and JAK-STAT pathways. Pharmacol Res. 2022 Aug;182:106347. doi: 10.1016/j.phrs.2022.106347[↩]
- Wiper A, Amoroso G, Rao S, Nolan J. Complex regional pain syndrome: a rare but potentially disabling complication of transradial cardiac catheterization. Catheter Cardiovasc Interv. 2020;95(5):E140–E143. doi: 10.1002/ccd.28357[↩]
- Crijns TJ, Van Der Gronde BATD, Ring D, Leung N. Complex regional pain syndrome after distal radius fracture is uncommon and is often associated with fibromyalgia. Clin Orthop Relat Res. 2018;476(4):744–750. doi: 10.1007/s11999.0000000000000070[↩][↩]
- Rochlin DH, Sheckter CC, Satteson ES, Swan CC, Fox PM, Curtin C. Separating fact from fiction: a nationwide longitudinal examination of complex regional pain syndrome following treatment of Dupuytren contracture. Hand. 2020 doi: 10.1177/1558944720963915[↩]
- Elsharydah A, Loo NH, Minhajuddin A, Kandil ES. Complex regional pain syndrome type 1 predictors—epidemiological perspective from a national database analysis. J Clin Anesth. 2017 doi: 10.1016/j.jclinane.2017.03.027[↩]
- Elsamadicy AA, Yang S, Sergesketter AR, et al. Prevalence and cost analysis of complex regional pain syndrome (CRPS): a role for neuromodulation. Neuromodulation. 2018;21(5):423–430. doi: 10.1111/ner.12691[↩]
- van Velzen GAJ, Huygen FJPM, van Kleef M, van Eijs FV, Marinus J, van Hilten JJ. Sex matters in complex regional pain syndrome. Eur J Pain (United Kingdom) 2019;23(6):1108–1116. doi: 10.1002/ejp.1375[↩][↩]
- Lipman MD, Hess DE, Werner BC, Deal DN. Fibromyalgia as a predictor of complex regional pain syndrome after distal radius fracture. Hand. 2019;14(4):516–522. doi: 10.1177/1558944717735949[↩]
- Birklein F, Ajit SK, Goebel A, Perez RSGM, Sommer C. Complex regional pain syndrome-phenotypic characteristics and potential biomarkers. Nat Rev Neurol. 2018;14(5):272–284. doi: 10.1038/nrneurol.2018.20[↩]
- Ward D, Thorsen NM, Frisch M, Valentiner-Branth P, Mølbak K, Hviid A. A cluster analysis of serious adverse event reports after human papillomavirus (HPV) vaccination in Danish girls and young women, September 2009 to August 2017. Euro Surveill. 2019;24(19):1. doi: 10.2807/1560-7917.ES.2019.24.19.1800380[↩]
- Hviid A, Thorsen NM, Valentiner-Branth P, Frisch M, Mølbak K. Association between quadrivalent human papillomavirus vaccination and selected syndromes with autonomic dysfunction in Danish females: population based, self-controlled, case series analysis. BMJ. 2020 doi: 10.1136/bmj.m2930[↩]
- Barboi A, Gibbons CH, Axelrod F, et al. Human papillomavirus (HPV) vaccine and autonomic disorders: a position statement from the American Autonomic Society. Auton Neurosci Basic Clin. 2020;223(1):13–18. doi: 10.1016/j.autneu.2019.05.002[↩]
- Pérez-Concha T, Tijero B, Acera M, et al. . Usefulness of thermography in the diagnosis and classification of complex regional pain syndrome. Neurología. 2020;38(5):342–349. doi: 10.1016/j.nrl.2020.10.011[↩]
- Castillo-Guzmán S, Nava-Obregón TA, Palacios-Ríos D, et al. . Complex regional pain syndrome (CRPS), a review. Medicina Universitaria. 2015;17(67):114–121. doi: 10.1016/j.rmu.2015.03.003[↩][↩][↩][↩]
- Lubkowska A, Pluta W.. Infrared thermography as a Non-Invasive tool in musculoskeletal disease rehabilitation–The control variables in applicability–A systematic review. Appl Sci. 2022;12(9):4302. doi: 10.3390/app12094302[↩]
- Kessler A, Yoo M, Calisoff R, et al. . Complex regional pain syndrome: an updated comprehensive review. NeuroRehabilitation. 2020;47(3):253–264. doi: 10.3233/NRE-208001[↩]
- Bruehl S, Harden RN, Galer BS, et al. . Complex regional pain syndrome: are there distinct subtypes and sequential stages of the syndrome? Pain. 2002;95(1-2):119–124. doi: 10.1016/s0304-3959(01)00387-6[↩]
- Bussa M, Guttilla D, Lucia M, Mascaro A, Rinaldi S. Complex regional pain syndrome type I: a comprehensive review. Acta Anaesthesiol Scand. 2015 Jul;59(6):685-97. doi: 10.1111/aas.12489[↩]
- Goebel A. Complex regional pain syndrome in adults. Rheumatology. 2011;50(10):1739-50. https://www.ncbi.nlm.nih.gov/pubmed/21712368[↩]
- O’Connell NE, Wand BM, McAuley J, Marston L, Moseley GL. Interventions for treating pain and disability in adults with complex regional pain syndrome. Cochrane Database Syst Rev. 2013 Apr 30;2013(4):CD009416. doi: 10.1002/14651858.CD009416.pub2. Update in: Cochrane Database Syst Rev. 2023 Jun 12;6:CD009416. doi: 10.1002/14651858.CD009416.pub3[↩]
- Duong S, Bravo D, Todd KJ, Finlayson RJ, Tran Q. Treatment of complex regional pain syndrome: an updated systematic review and narrative synthesis. Can J Anaesth. 2018 Jun;65(6):658-684. English. doi: 10.1007/s12630-018-1091-5[↩]
- Mangnus TJP, Dirckx M, Huygen FJPM. Different Types of Pain in Complex Regional Pain Syndrome Require a Personalized Treatment Strategy. J Pain Res. 2023 Dec 27;16:4379-4391. doi: 10.2147/JPR.S432209[↩]
- Shim H, Rose J, Halle S, et al. . Complex regional pain syndrome: a narrative review for the practising clinician. Br J Anaesth. 2019;123(2):e424–e433. doi: 10.1016/J.BJA.2019.03.03.0[↩][↩][↩]
- Collins CK. Physical therapy management of complex regional pain syndrome I in a 14-year-old patient using strain counterstrain: a case report. J Man Manip Ther. 2007;15(1):25–41 doi: 10.1179/106698107791090150[↩]
- Menck JY, Requejo SM, Kulig K.. Thoracic spine dysfunction in upper extremity complex regional pain syndrome type I. J Orthop Sports Phys Ther. 2000;30(7):401–409. doi: 10.2519/JOSPT.2000.30.7.401[↩]
- Cleland J, McRae M.. Complex regional pain syndrome I: management through the use of vertebral and sympathetic trunk mobilization. J Man Manip Ther. 2002;10(4):188–199. doi: 10.1179/106698102790819067[↩]
- Gutiérrez-Espinoza H, Tabach-Apraiz A, Oyanadel-Maldonado M.. Physical therapy in patients with complex regional pain syndrome type I after distal radius fracture: a case series. J Phys Ther Sci. 2019;31(4):403–407. doi: 10.1589/JPTS.31.40.3[↩]
- Walston Z, Hernandez L, Yake D.. Utilization of manual therapy to the lumbar spine in conjunction with traditional conservative care for individuals with bilateral lower extremity complex regional pain syndrome: A case series. Physiother Theory Pract. 2018;36(1):241–248. doi: 10.1080/09593985.2018.1482392[↩]
- Lohnberg JA, Altmaier EM.. A review of psychosocial factors in complex regional pain syndrome. J Clin Psychol Med Settings. 2013;20(2):247–254. doi: 10.1007/S10880-012-9322-3[↩]
- Edwards RR, Smith MT, Kudel I, et al. . Pain-related catastrophizing as a risk factor for suicidal ideation in chronic pain. Pain. 2006;126(1-3):272–279. doi: 10.1016/J.PAIN.2006.07.004[↩]
- Bodde MI, Schrier E, Krans HK, et al. . Resilience in patients with amputation because of complex regional pain syndrome type I. Disabil Rehabil. 2014;36(10):838–843. doi: 10.3109/09638288.2013.822023[↩][↩]
- Tang NKY, Crane C.. Suicidality in chronic pain: a review of the prevalence, risk factors and psychological links. Psychol Med. 2006;36(5):575–586. doi: 10.1017/S0033291705006859[↩]
- Ryskalin L, Ghelarducci G, Marinelli C, et al. . Effectiveness of decision support to treat complex regional pain syndrome. Appl Sci. 2022;12(18):8979. doi: 10.3390/app12188979[↩]
- Fialka V, Korpan M, Saradeth T, et al. . Autogenic training for reflex sympathetic dystrophy: a pilot study. Complement Ther Med. 1996;4(2):103–105. doi: 10.1016/S0965-2299(96)80026-4[↩][↩]
- Albazaz R, Wong YT, Homer-Vanniasinkam S.. Complex regional pain syndrome: a review. Ann Vasc Surg. 2008;22(2):297–306. doi: 10.1016/J.AVSG.2007.10.006[↩]
- Mackey S, Feinberg S.. Pharmacologic therapies for complex regional pain syndrome. Curr Pain Headache Rep. 2007;11(1):38–43. doi: 10.1007/S11916-007-0020-Z[↩]
- Nazir SST, Ivan N, Antonella U, et al. . Complex regional pain syndrome: a comprehensive review. Pain Ther. 2021;10(2):875–892. doi: 10.1007/S40122-021-00279-4[↩]
- Resmini G, Ratti C, Canton G, et al. . Treatment of complex regional pain syndrome. Clin Cases Miner Bone Metab. 2015;12(Suppl 1):26–30. doi: 10.11138/CCMBM/2015.12.3S.026[↩]
- Kalita J, Vajpayee A, Misra UK.. Comparison of prednisolone with piroxicam in complex regional pain syndrome following stroke: a randomized controlled trial. QJM. 2006;99(2):89–95. doi: 10.1093/QJMED/HCL004[↩]
- Atalay N, Ercidogan O, Akkaya N, et al. . Prednisolone in complex regional pain syndrome. Pain Phys. 2014;2;17(2;3):179–185. doi: 10.36076/ppj.2014/17/179[↩]
- Bianchi C, Rossi S, Turi S, Brambilla A, Felisari G, Mascheri D. Long-term functional outcome measures in corticosteroid-treated complex regional pain syndrome. Eura Medicophys. 2006 Jun;42(2):103-11. https://www.minervamedica.it/en/journals/europa-medicophysica/article.php?cod=R33Y2006N02A0103[↩]
- Jamroz A, Berger M, Winston P.. Prednisone for acute complex regional pain syndrome: a retrospective cohort study. Pain Res Manag. 2020;2020:8182569. doi: 10.1155/2020/8182569[↩]
- Gelman B, Hsu ES.. Gabapentin. In: Sinatra RS, Jahr JS, J. Watkins-Pitchford M (editors). The essence of analgesia and analgesics. Cambridge: Cambridge University Press; 2022. p. 294–297. doi: 10.1017/CBO9780511841378.071[↩]
- van de Vusse AC, Stomp-van den Berg SGM, Kessels AHF, et al. . Randomised controlled trial of gabapentin in complex regional pain syndrome type 1 [ISRCTN84121379]. BMC Neurol. 2004;4(1):13. doi: 10.1186/1471-2377-4-13[↩]
- Brown S, Johnston B, Amaria K, et al. . A randomized controlled trial of amitriptyline versus gabapentin for complex regional pain syndrome type I and neuropathic pain in children. Scand J Pain. 2016;13(1):156–163. doi: 10.1016/J.SJPAIN.2016.05.039[↩]
- Varenna M, Adami S, Sinigaglia L. Bisphosphonates in Complex Regional Pain syndrome type I: how do they work? Clin Exp Rheumatol. 2014 Jul-Aug;32(4):451-4. https://www.clinexprheumatol.org/abstract.asp?a=8025[↩][↩]
- Chevreau M, Romand X, Gaudin P, et al. . Bisphosphonates for treatment of complex regional pain syndrome type 1: a systematic literature review and meta-analysis of randomized controlled trials versus placebo. Joint Bone Spine. 2017;84(4):393–399. doi: 10.1016/J.JBSPIN.2017.03.009[↩]
- Connolly SB, Prager JP, Harden RN.. A systematic review of ketamine for complex regional pain syndrome. Pain Med. 2015;16(5):943–969. doi: 10.1111/PME.12675[↩][↩]
- Rosenbaum SB, Gupta V, Patel P, et al. Ketamine. [Updated 2024 Jan 30]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK470357[↩]
- Azari P, Lindsay DR, Briones D, et al. . Efficacy and safety of ketamine in patients with complex regional pain syndrome. CNS Drugs. 2012;26(3):215–228. doi: 10.2165/11595200-000000000-00000[↩]
- Urits I, Shen AH, Jones MR, Viswanath O, Kaye AD. Complex Regional Pain Syndrome, Current Concepts and Treatment Options. Curr Pain Headache Rep. 2018 Feb 5;22(2):10. doi: 10.1007/s11916-018-0667-7[↩]
- Wissel J, Müller J, Dressnandt J, et al. . Management of spasticity associated pain with botulinum toxin A. J Pain Symptom Manage. 2000;20(1):44–49. doi: 10.1016/S0885-3924(00)00146-9[↩]
- Schilder JC, van Dijk JG, Dressler D, Koelman JH, Marinus J, van Hilten JJ. Responsiveness to botulinum toxin type A in muscles of complex regional pain patients with tonic dystonia. J Neural Transm (Vienna). 2014 Jul;121(7):761-7. doi: 10.1007/s00702-014-1172-8[↩]
- Carroll I, Clark JD, Mackey S.. Sympathetic block with botulinum toxin to treat complex regional pain syndrome. Ann Neurol. 2009;65(3):348–351. doi: 10.1002/ANA.21601[↩]
- Siongco PRL, Rosales RL, Moore AP, Freynhagen R, Arimura K, Kanovsky P, Kaji R, Fernandez HH, Dressler D. Botulinum neurotoxin injections for muscle-based (dystonia and spasticity) and non-muscle-based (neuropathic pain) pain disorders: a meta-analytic study. J Neural Transm (Vienna). 2020 Jun;127(6):935-951. doi: 10.1007/s00702-020-02163-5[↩]
- Lee Y, Lee CJ, Choi E, et al. . Lumbar sympathetic block with botulinum toxin type a and type B for the complex regional pain syndrome. Toxins. 2018;10(4):164. doi: 10.3390/TOXINS10040164[↩]
- Comella CL, Jankovic J, Shannon KM, et al. . Comparison of botulinum toxin serotypes a and B for the treatment of cervical dystonia. Neurology. 2005;65(9):1423–1429. doi: 10.1212/01.WNL.0000183055.81056.5C[↩]
- Tokuno H. Efficacy of Botulinum toxin injection in reducing limb pain in patients with complex regional pain syndrome. https://clinicaltrials.gov/study/NCT03616262[↩]
- Singh V, Cohen SP.. Prolonging sympathetic blockade for complex regional pain syndrome: is botulinum toxin the answer? Anesthesiology. 2022;136(2):261–264. doi: 10.1097/ALN.0000000000004093[↩]
- Eisenberg E, Shtahl S, Geller R, et al. . Serum and salivary oxidative analysis in complex regional pain syndrome. Pain. 2008;138(1):226–232. doi: 10.1016/J.PAIN.2008.04.019[↩]
- Baykal T, Seferoglu B, Karsan O, et al. . Antioxidant profile in patients with complex regional pain syndrome type I. Int J Rheum Dis. 2014;17(2):156–158. doi: 10.1111/1756-185X.12140[↩]
- Hernigou J, Labadens A, Ghistelinck B, Bui Quoc E, Maes R, Bhogal H, Callewier A, Bath O, Chahidi E, Safar A. Vitamin C prevention of complex regional pain syndrome after foot and ankle surgery: a prospective randomized study of three hundred and twenty nine patients. Int Orthop. 2021 Sep;45(9):2453-2459. doi: 10.1007/s00264-021-05159-2[↩]
- Jacques H, Jérôme V, Antoine C, Lucile S, Valérie D, Amandine L, Theofylaktos K, Olivier B. Prospective randomized study of the vitamin C effect on pain and complex pain regional syndrome after total knee arthroplasty. Int Orthop. 2021 May;45(5):1155-1162. doi: 10.1007/s00264-020-04936-9[↩]
- Laumonerie P, Martel M, Tibbo ME, Azoulay V, Mansat P, Bonnevialle N. Influence of vitamin C on the incidence of CRPS-I after subacromial shoulder surgery. Eur J Orthop Surg Traumatol. 2020 Feb;30(2):221-226. doi: 10.1007/s00590-019-02542-z[↩]
- Aïm F, Klouche S, Frison A, et al. . Efficacy of vitamin C in preventing complex regional pain syndrome after wrist fracture: a systematic review and meta-analysis. Orthop Traumatol Surg Res. 2017;103(3):465–470. doi: 10.1016/J.OTSR.2016.12.021[↩]
- O’Connell NE, Wand BM, Gibson W, Carr DB, Birklein F, Stanton TR. Local anaesthetic sympathetic blockade for complex regional pain syndrome. Cochrane Database Syst Rev. 2016 Jul 28;7(7):CD004598. doi: 10.1002/14651858.CD004598.pub4[↩][↩]
- Cheng J, Salmasi V, You J, et al. . Outcomes of sympathetic blocks in the management of complex regional pain syndrome: a retrospective cohort study. Anesthesiology. 2019;131(4):883–893. doi: 10.1097/ALN.0000000000002899[↩]
- Visnjevac O, Costandi S, Patel BA, Azer G, Agarwal P, Bolash R, Mekhail NA. A Comprehensive Outcome-Specific Review of the Use of Spinal Cord Stimulation for Complex Regional Pain Syndrome. Pain Pract. 2017 Apr;17(4):533-545. doi: 10.1111/papr.12513[↩][↩][↩]
- Hoikkanen T, Nissen M, Ikäheimo TM, Jyrkkänen HK, Huttunen J, von Und Zu Fraunberg M. Long-Term Outcome of Spinal Cord Stimulation in Complex Regional Pain Syndrome. Neurosurgery. 2021 Sep 15;89(4):597-609. doi: 10.1093/neuros/nyab239[↩][↩]
- Graham RD, Sankarasubramanian V, Lempka SF.. Dorsal root ganglion stimulation for chronic pain: hypothesized mechanisms of action. J Pain. 2022;23(2):196–211. doi: 10.1016/J.JPAIN.2021.07.008[↩]
- Koetsier E, Franken G, Debets J, et al. . Mechanism of dorsal root ganglion stimulation for pain relief in painful diabetic polyneuropathy is not dependent on GABA release in the dorsal horn of the spinal cord. CNS Neurosci Ther. 2020;26(1):136–143. doi: 10.1111/CNS.13192[↩]
- Deer TR, Levy RM, Kramer J, et al. . Dorsal root ganglion stimulation yielded higher treatment success rate for complex regional pain syndrome and causalgia at 3 and 12 months: a randomized comparative trial. Pain. 2017;158(4):669–681. doi: 10.1097/J.PAIN.0000000000000814[↩][↩][↩]
- Koopmeiners AS, Mueller S, Kramer J, et al. . Effect of electrical field stimulation on dorsal root ganglion neuronal function. Neuromodulation. 2013;16(4):304–311. doi: 10.1111/NER.12028[↩]
- Huygen FJPM, Liem L, Nijhuis H, et al. . Evaluating dorsal root ganglion stimulation in a prospective Dutch cohort. Neuromodulation. 2019;22(1):80–86. doi: 10.1111/NER.12798[↩]
- Rauck RL, Eisenach JC, Jackson K, Young LD, Southern J. Epidural clonidine treatment for refractory reflex sympathetic dystrophy. Anesthesiology. 1993 Dec;79(6):1163-9; discussion 27A.[↩][↩][↩][↩][↩][↩]
- Cooper DE, DeLee JC, Ramamurthy S. Reflex sympathetic dystrophy of the knee. Treatment using continuous epidural anesthesia. J Bone Joint Surg Am. 1989 Mar;71(3):365-9. https://journals.lww.com/jbjsjournal/abstract/1989/71030/reflex_sympathetic_dystrophy_of_the_knee_.9.aspx[↩][↩]
- Koning H, Christiaans C, Overdijk G, Mackie D.. Cervical epidural blockade and reflex sympathetic dystrophy. Pain Clinic 1995;8:239–44.[↩]
- Buchheit T, Crews JC. Lateral cervical epidural catheter placement for continuous unilateral upper extremity analgesia and sympathetic block. Reg Anesth Pain Med. 2000 May-Jun;25(3):313-7. doi: 10.1016/s1098-7339(00)90019-1[↩]
- Du Pen SL, Peterson DG, Williams A, Bogosian AJ. Infection during chronic epidural catheterization: diagnosis and treatment. Anesthesiology. 1990 Nov;73(5):905-9. doi: 10.1097/00000542-199011000-00018[↩][↩]
- Lundborg C, Dahm P, Nitescu P, Appelgren L, Curelaru I. Clinical experience using intrathecal (IT) bupivacaine infusion in three patients with complex regional pain syndrome type I (CRPS-I). Acta Anaesthesiol Scand. 1999 Jul;43(6):667-78. doi: 10.1034/j.1399-6576.1999.430613.x[↩][↩]
- van Hilten BJ, van de Beek WJ, Hoff JI, Voormolen JH, Delhaas EM. Intrathecal baclofen for the treatment of dystonia in patients with reflex sympathetic dystrophy. N Engl J Med. 2000 Aug 31;343(9):625-30. doi: 10.1056/NEJM200008313430905[↩][↩][↩]
- Attal N, Brasseur L, Chauvin M, Bouhassira D. Effects of single and repeated applications of a eutectic mixture of local anaesthetics (EMLA) cream on spontaneous and evoked pain in post-herpetic neuralgia. Pain. 1999 May;81(1-2):203-9. doi: 10.1016/s0304-3959(99)00014-7[↩]
- Galer BS, Rowbotham MC, Perander J, Friedman E. Topical lidocaine patch relieves postherpetic neuralgia more effectively than a vehicle topical patch: results of an enriched enrollment study. Pain. 1999 Apr;80(3):533-538. doi: 10.1016/S0304-3959(98)00244-9[↩]
- Rowbotham MC, Davies PS, Verkempinck C, Galer BS. Lidocaine patch: double-blind controlled study of a new treatment method for post-herpetic neuralgia. Pain. 1996 Apr;65(1):39-44. doi: 10.1016/0304-3959(95)00146-8[↩]
- Chang A, Rosani A, Quick J. Capsaicin. [Updated 2023 May 23]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK459168[↩][↩]
- Watson CP, Tyler KL, Bickers DR, Millikan LE, Smith S, Coleman E. A randomized vehicle-controlled trial of topical capsaicin in the treatment of postherpetic neuralgia. Clin Ther. 1993 May-Jun;15(3):510-26.[↩]
- Robbins WR, Staats PS, Levine J, Fields HL, Allen RW, Campbell JN, Pappagallo M. Treatment of intractable pain with topical large-dose capsaicin: preliminary report. Anesth Analg. 1998 Mar;86(3):579-83. doi: 10.1097/00000539-199803000-00027[↩]
- Bernstein JE, Korman NJ, Bickers DR, Dahl MV, Millikan LE. Topical capsaicin treatment of chronic postherpetic neuralgia. J Am Acad Dermatol. 1989 Aug;21(2 Pt 1):265-70. doi: 10.1016/s0190-9622(89)70171-7[↩]
- Treatment of painful diabetic neuropathy with topical capsaicin. A multicenter, double-blind, vehicle-controlled study. The Capsaicin Study Group. Arch Intern Med. 1991 Nov;151(11):2225-9. doi: 10.1001/archinte.151.11.2225[↩]
- Peikert A, Hentrich M, Ochs G. Topical 0.025% capsaicin in chronic post-herpetic neuralgia: efficacy, predictors of response and long-term course. J Neurol. 1991 Dec;238(8):452-6. doi: 10.1007/BF00314653[↩]
- Simone DA, Ochoa J. Early and late effects of prolonged topical capsaicin on cutaneous sensibility and neurogenic vasodilatation in humans. Pain. 1991 Dec;47(3):285-294. doi: 10.1016/0304-3959(91)90217-L[↩]
- Flor H, Fydrich T, Turk DC. Efficacy of multidisciplinary pain treatment centers: a meta-analytic review. Pain. 1992 May;49(2):221-230. doi: 10.1016/0304-3959(92)90145-2[↩]
- Backonja M, Wallace MS, Blonsky ER, Cutler BJ, Malan P Jr, Rauck R, Tobias J; NGX-4010 C116 Study Group. NGX-4010, a high-concentration capsaicin patch, for the treatment of postherpetic neuralgia: a randomised, double-blind study. Lancet Neurol. 2008 Dec;7(12):1106-12. doi: 10.1016/S1474-4422(08)70228-X Erratum in: Lancet Neurol. 2009 Jan;8(1):31.[↩]
- Simpson DM, Brown S, Tobias J; NGX-4010 C107 Study Group. Controlled trial of high-concentration capsaicin patch for treatment of painful HIV neuropathy. Neurology. 2008 Jun 10;70(24):2305-13. doi: 10.1212/01.wnl.0000314647.35825.9c[↩]
- Simpson DM, Gazda S, Brown S, Webster LR, Lu SP, Tobias JK, Vanhove GF; NGX-4010 C118 Study Group. Long-term safety of NGX-4010, a high-concentration capsaicin patch, in patients with peripheral neuropathic pain. J Pain Symptom Manage. 2010 Jun;39(6):1053-64. doi: 10.1016/j.jpainsymman.2009.11.316[↩]
- Girtler R, Kloimstein H, Gustorff B. Ausgeprägte Symptomverschlechterung bei CRPS Typ II nach einmaliger Applikation eines hochprozentigen Capsaicinpflasters. Ein Fallbericht [Pronounced symptom deterioration in complex regional pain syndrome type II after isolated application of a highly concentrated capsaicin patch. A case report]. Schmerz. 2013 Feb;27(1):67-71. German. doi: 10.1007/s00482-012-1268-8[↩]
- Zuurmond WW, Langendijk PN, Bezemer PD, Brink HE, de Lange JJ, van loenen AC. Treatment of acute reflex sympathetic dystrophy with DMSO 50% in a fatty cream. Acta Anaesthesiol Scand. 1996 Mar;40(3):364-7. doi: 10.1111/j.1399-6576.1996.tb04446.x[↩]
- Forouzanfar T, Köke AJ, van Kleef M, Weber WE. Treatment of complex regional pain syndrome type I. Eur J Pain. 2002;6(2):105-22. doi: 10.1053/eujp.2001.0304[↩]
- Goebel A, Baranowski A, Maurer K, Ghiai A, McCabe C, Ambler G. Intravenous immunoglobulin treatment of the complex regional pain syndrome: a randomized trial. Ann Intern Med. 2010 Feb 2;152(3):152-8. doi: 10.7326/0003-4819-152-3-201002020-00006[↩]
- Goebel A, Bisla J, Carganillo R, Frank B, Gupta R, Kelly J, McCabe C, Murphy C, Padfield N, Phillips C, Sanders M, Serpell M, Shenker N, Shoukrey K, Wyatt L, Ambler G. Low-Dose Intravenous Immunoglobulin Treatment for Long-Standing Complex Regional Pain Syndrome: A Randomized Trial. Ann Intern Med. 2017 Oct 3;167(7):476-483. doi: 10.7326/M17-0509[↩]
- Aradillas E, Schwartzman RJ, Grothusen JR, Goebel A, Alexander GM. Plasma Exchange Therapy in Patients with Complex Regional Pain Syndrome. Pain Physician. 2015 Jul-Aug;18(4):383-94. https://www.painphysicianjournal.com/linkout?issn=&vol=18&page=383[↩]
- Schwartz J, Padmanabhan A, Aqui N, Balogun RA, Connelly-Smith L, Delaney M, Dunbar NM, Witt V, Wu Y, Shaz BH. Guidelines on the Use of Therapeutic Apheresis in Clinical Practice-Evidence-Based Approach from the Writing Committee of the American Society for Apheresis: The Seventh Special Issue. J Clin Apher. 2016 Jun;31(3):149-62. doi: 10.1002/jca.21470[↩]
- Krans-Schreuder HK, Bodde MI, Schrier E, et al. . Amputation for long-standing, therapy-resistant type-I complex regional pain syndrome. J Bone Joint Surg Am. 2012;94(24):2263–2268. doi: 10.2106/JBJS.L.00532[↩]
- Honkamp N, Amendola A, Hurwitz S, et al. . Retrospective review of eighteen patients who underwent transtibial amputation for intractable pain. J Bone Joint Surg Am. 2001;83(10):1479–1483. doi: 10.2106/00004623-200110000-00003[↩]
- Kashy BK, Abd-Elsayed AA, Farag E, et al. . Amputation as an unusual treatment for Therapy-Resistant complex regional pain syndrome, type 1. Ochsner J. 2015;15(4):441–442. doi: 10.1043/TOJ-14-0074[↩]