Evidence-Based Pharmacotherapy of Chronic Pain



Fig. 28.1
Steps in urine drug testing in chronic pain








Antidepressants



Tricyclic Antidepressants


Antidepressants are a heterogeneous group of drugs which have all demonstrated beneficial activity for patients with major depressive order. Additionally, these agents may have beneficial effects in patients with anxiety disorders (e.g. panic disorder, generalized anxiety disorder, social anxiety, post-traumatic stress disorder, and obsessive–compulsive disorder), eating disorders, attention deficit hyperactivity disorders, premenstrual dysphoric disorder, and chronic pain. Many antidepressants possess analgesic qualities for a broad range of pain conditions independent of their effects on mood.

Antidepressant medications may be classified as follows (Smith 2007):

1.

Cyclic antidepressants, including the TCAs and tetracyclic antidepressants (e.g., maprotiline)

 

2.

Selective serotonin reuptake inhibitors (SSRIs)

 

3.

Serotonin norepinephrine reuptake inhibitors (SNRIs)

 

4.

Dopamine norepinephrine reuptake inhibitors (DNRIs)

 

5.

Norepinephrine reuptake inhibitors (NRIs)

 

6.

Monoamine oxidase inhibitors (MAOIs)

 

7.

The miscellaneous category of “atypical antidepressants”

 

Although the role of SSRIs in providing effective analgesia is uncertain, it appears limited at best (Saarto and Wiffen 2007). Two classes of antidepressants have significant analgesic properties, the TCAs, and SNRIs. Both TCAs and SNRIs (e.g., venlafaxine) have NNT roughly in the neighborhood of three (Saarto and Wiffen 2007).

Most potent effects on the noradrenergic system is desipramine [also going along with it being least sedating] (nortriptyline also exhibits preferential inhibition of NE reuptake over 5-HT reuptake). Doxepin has the most potent anti-histaminergic effects.

The TCAs can be divided into amines and their demethylated secondary amine derivatives. In addition, maprotiline (Ludiomil) is classified as a tetracyclic.

The tertiary amine TCAs include the following:



  • Amitriptyline (Elavil)


  • Imipramine (Tofranil)


  • Trimipramine (Surmontil)


  • Clomipramine (Anafranil)


  • Doxepin (Sinequan)

The secondary amine TCAs include the following:



  • Nortriptyline (Pamelor)


  • Desipramine (Norpramin)


  • Protriptyline (Vivactil)


  • Amoxapine (Asendin)

TCAs, although having reasonable analgesic properties, may also possess multiple undesirable adverse effects. These adverse effects may vary depending on the individual agent. Among the TCAs, amitriptyline has the most potent anticholinergic effects and desipramine has the least, and thus is the least sedating.

Multiple placebo-controlled RCTs have found TCAs to be efficacious for several different types of neuropathic pain (Dworkin et al. 2007; Finnerup et al. 2005, 2007, 2010; Saarto and Wiffen 2007; Sindrup et al. 2005). In addition, TCAs are efficacious for the treatment of depression, a common comorbidity in patients with chronic pain, but the analgesic efficacy in neuropathic pain has been established in nondepressed patients, which establishes their beneficial effect in neuropathic pain cannot be explained simply by antidepressant effects. Anticholinergic adverse effects are common and include dry mouth, orthostatic hypotension, constipation, and urinary retention. These effects can be reduced by starting with low dosages administered at bedtime and with slow titration to higher dosage as well as by using a secondary amine TCAs (e.g., nortriptyline or desipramine).

The Neuropathic Special Interest Group (NeuPSIG) of the International Association for the Study of Pain (IASP) developed guidelines which recommend prescribing TCAs, with caution in patients with ischemic cardiac disease or ventricular conduction abnormalities, limiting the dosages to less than 100 mg/day when possible and obtaining a screening electrocardiogram for patients older than 40 years (Dworkin et al. 2007).

It can take up to 6–8 weeks, including 2 weeks at the highest dosage tolerated, for an adequate trial of treatment with TCA however, analgesia tends to be evident quicker (after about a week) and at lower does than when these agents are utilized to treat depression (Dworkin et al. 2007; Finnerup et al. 2005).


Antidepressants with Both Norepinephrine and Serotonin Reuptake Inhibition


There are four SNRIs available in the United States. Venlafaxine (Effexor) and Desvenlafaxine (Pristiq) are FDA-approved for the treatment of major depressive disorder. Milnacipran is FDA-approved for the treatment of fibromyalgia. Duloxetine and venlafaxine are selective SNRIs that have been studied in peripheral neuropathic pain (another SNRI, milnacipran has been studied only in fibromyalgia).

Venlafaxine has shown efficacy in painful diabetic peripheral neuropathy and painful polyneuropathies of different origins, but not in postherpetic neuralgia (Dworkin et al. 2007). Typically 2–4 weeks are required to titrate to an efficacious dosage (that is 150–225 mg/day); venlafaxine is available in short- and long-acting preparations. At very low doses, venlafaxine tends to “act” more like an SSRI but is clearly an SNRI as the dose is increased. Cardiac conduction abnormalities have been reported in a small number of patients, and blood pressure increases can occur; therefore venlafaxine should be prescribed with caution in patients with cardiac disease. In addition, venlafaxine should be tapered when treatment is being discontinued because of withdrawal symptoms.

The U.S. Food and Drug Administration approved duloxetine hydrochloride to treat chronic musculoskeletal pain on November 4, 2010, including discomfort from osteoarthritis and chronic lower back pain. Cymbalta was first used to treat major depressive disorder in 2004. Since its initial approval, over 30 million patients in the United States have used duloxetine. It was approved for the treatment of diabetic peripheral neuropathy in 2004; generalized anxiety disorder and maintenance treatment of major depression in 2007; and fibromyalgia in 2008. More than 29,000 patients have used duloxetine in clinical trials, and more than 600 patients were studied in the clinical trials involving osteoarthritis and CLBP. The safety evaluation for duloxetine included review of data from the clinical trials as well as postmarketing data from the previously approved patient populations. The FDA assessed the efficacy of duloxetine in CLBP and osteoarthritis in four double-blind, placebo-controlled, randomized clinical trials. At the end of the study period, patients taking duloxetine had a significantly greater pain reduction compared with placebo. The most common side effects reported with duloxetine include nausea, dry mouth, insomnia, drowsiness, constipation, fatigue, and dizziness. Other serious side effects include liver damage, allergic reactions such as hives, rashes and/or swelling of the face, pneumonia, depressed mood, suicide, suicidal thoughts and behavior.

Skljarevski et al. (2010) conducted a randomized, double-blind, placebo-controlled study; and assessed efficacy and safety of duloxetine in patients with CLBP. Adults (n  =  401) with a nonneuropathic CLBP and average pain intensity of ≥4 on an 11-point numerical scale (Brief Pain Inventory [BPI]) were treated with either duloxetine 60 mg once daily or placebo for 12 weeks. The primary measure was BPI average pain (Skljarevski et al. 2010). Compared with placebo-treated patients, duloxetine-treated patients reported a significantly greater reduction in BPI average pain (p  ≤  0.001). Similarly, duloxetine-treated patients reported significantly greater improvements in PGI-I, BPI-S, BPI-I, 50% response rates, and some health outcomes. The RMDQ and 30% response rate showed numerical improvements with duloxetine treatment. Significantly more patients in the duloxetine group (15.2%) than patients in the placebo group (5.4%) discontinued because of adverse events (p  =  0.002). Nausea and dry mouth were the most common treatment-emergent adverse events with rates significantly higher in duloxetine-treated patients (Skljarevski et al. 2010).

Duloxetine has shown consistent efficacy in painful polyneuropathy (DPN) (Dworkin et al. 2007) with effectiveness sustained for 1 year in an open label trial (Raskin et al. 2006). Unfortunately, Duloxetine has not been studied in any other types of neuropathic pain and so its efficacy in such conditions is unknown. The most common adverse affect of Duloxetine is nausea, which seems to be reduced by administering 30 mg once daily for 1 week before increasing to 60 mg once daily. Duloxetine does not seem to produce clinically important electrocardiographic or blood pressure changes and our recent review concluded that aminotransferase monitoring is unnecessary.


Calcium Channel Alpha 2-Delta Ligands (Gabapentin and Pregabalin)


Gabapentin and pregabalin each bind to the voltage-gated calcium channels at the alpha 2-delta subunit and inhibit neurotransmitter release. They have shown efficacy vs. placebo in several neuropathic pain conditions (Dworkin et al. 2007; Finnerup et al. 2005; Finnerup and Jensen 2007; Finnerup 2010). Although gabapentin and pregabalin have few drug interactions, both can produce dose-dependent dizziness and sedation, which can be reduced by starting with lower dosages and titrating cautiously. Both medications also require dosage reduction in patients with renal insufficiency and dosage adjustments can be made in relation to creatinine clearance.

Gabapentin is absorbed slowly after oral administration, with maximum plasma concentrations attained within 3–4 h (Bockbrader et al. 2010). Orally administered gabapentin exhibits saturable absorption – a nonlinear (zero-order) process – making its pharmacokinetics less predictable. Plasma concentrations of gabapentin do not increase proportionally with increasing dose (Bockbrader et al. 2010). Orally administered pregabalin is absorbed more rapidly, with maximum plasma concentrations attained within 1 h. Absorption is linear (first order), with plasma concentrations increasing proportionately with increasing dose. The absolute bioavailability of gabapentin drops from 60 to 33% as the dosage increases from 900 to 3,600 mg/day, while the absolute bioavailability of pregabalin remains at ≥90% irrespective of the dosage (Bockbrader et al. 2010). Treatment should be initiated at low dosage with gradual increases until pain relief, dose-limiting adverse affects, or 3,600 mg/day in three divided doses reached. An adequate trial of treatment with gabapentin can require 2 months or more.

A systematic review of 15 trials (1,468 participants) evaluating gabapentin included 1 acute pain trial and 14 trials in neuropathic (7 in diabetic neuropathy, 2 in postherpetic neuralgia and 1 each in cancer-related neuropathy, phantom limb pain, spinal cord injury, Guillain–Barre syndrome and miscellaneous neuropathies) (Wiffen et al. 2005). In the 14 chronic neuropathic pain trials, 42% of the participants improved on gabapentin (that is pain relief of 50% or greater on gabapentin vs. 19% on placebo, and the NNT for improvement in all trials with evaluable data was 4.3 (95% CI, 3.5–5.7)). Withdrawal rates were 14% for gabapentin vs. 10% for placebo.

In an updated Cochrane Review evaluating gabapentin for chronic neuropathic pain and fibromyalgia in adults, Moore et al. (2011) found that gabapentin provides pain relief of a high level in about a third of people who take it for painful neuropathic pain. Using the Initiative on Methods, Measurement and Pain Assessment in Clinical Trials (IMMPACT) definition of at least moderate benefit, gabapentin was superior to placebo in 14 studies with 2,831 participants, 43% improving with gabapentin and 26% with placebo; the NNT was 5.8 (4.8–7.2) (Moore et al. 2011). Using the IMMPACT definition of substantial benefit, gabapentin was superior to placebo in 13 studies with 2,627 participants, 31% improving with gabapentin and 17% with placebo; the NNT was 6.8 (5.6–8.7) (Moore et al. 2011).

Pregabalin may provide analgesia more quickly then gabapentin because initial dosage of 150 mg/day has been found to be efficacious in some trials and because of the time required to titrate to a full dosage is less (Stacey et al. 2008).

Moore et al. (2009) performed a Cochrane Review of pregabalin for acute and chronic pain. For chronic pain, pregabalin at 150 mg daily was generally ineffective. Efficacy was demonstrated for dichotomous outcomes equating to moderate or substantial pain relief, alongside lower rates for lack of efficacy discontinuations with increasing dose. The best (lowest) NNT for each condition for at least 50% pain relief over baseline (substantial benefit) for 600 mg pregabalin daily compared with placebo were 3.9 (95% confidence interval 3.1–5.1) for postherpetic neuralgia, 5.0 (4.0–6.6) for painful diabetic neuropathy, 5.6 (3.5–14) for central neuropathic pain, and 11 (7.1–21) for fibromyalgia (Moore et al. 2009). The FDA has improved pregabalin for treatment of neuropathic pain associated with diabetic peripheral neuropathy and postherpetic neuralgia and for treatment of fibromyalgia; evidence from these trials is discussed under specific disorders.


Skeletal Muscle Relaxants


Most muscle relaxants are FDA-approved for either spasticity (baclofen, dantrolene, and tizanidine) or musculoskeletal conditions (carisoprodol, chlorzoxazone, cyclobenzaprine, metaxalone, methocarbamol, or orphenadrine) (Chou et al. 2004). The mechanism of action for the latter category of agents is unclear, but may be related in part to sedative effects. Cyclobenzaprine is structurally very similar to amitriptyline and may have similar mechanisms of action. Studies to date have not shown differences amongst most skeletal muscle relaxants with respect to their efficacy, adverse affects or safety. A systemic review (Chou et al. 2004) concluded that there was “insufficient data to assess comparative abuse and addiction of skeletal muscle relaxants of almost all case reports an abuse and addiction have been in patients taking carisoprodol.” Most trials are focused on acute rather than chronic pain. Cyclobenzaprine is the best studied of muscle relaxant in musculoskeletal disorders overall; in 21 fair quality trials it is consistently proven superior to placebo for fibromyalgia, as well as for pain relief, muscle spasm and functional status in other disorders. Cyclobenzaprine 5 mg t.i.d. tends to be as equally effective as 10 mg t.i.d. but has fewer side effects.


Topical Analgesics



Lidocaine 5% Patch


The 5% lidocaine patch has shown efficacy and excellent tolerability in trials involving patients with postherpetic neuralgia and allodynia and in patients with allodynia due to different types of peripheral neuropathic pain (Davies and Galer 2004; Khaliq et al. 2007). As a topical treatment without substantial systemic absorption, the most common adverse effects are mild local reactions. The lack of systemic adverse affects and drug interactions can be particularly advantageous in all the patients or patients with complex neuropathic pain. Lidocaine gel (5%), which is less expensive than the lidocaine patch, has also shown efficacy in patients with postherpetic neuralgia and allodynia. Topical lidocaine is most appropriate in well-localized neuropathic pain and is likely to be of benefit in patients with central neuropathic pain. Unfortunately, attempts to predict which patients are most likely to respond to this treatment with topical lidocaine have been generally unsuccessful.


Capsaicin


Capsaicin is an alkaloid derived from chili peppers; repeated application is thought to lead to depletion of substance from primary afferent neurons (Lee et al. 1991). The main disadvantage of capsaicin is the initial burning sensation, which may persist for days. Capsaicin must be applied 3–4 times per day over the entire painful area for up to 6–8 weeks before optimal pain relief can be achieved. Mason et al. (2004a) reviewed the clinical trial evidence for capsaicin, including six trials in neuropathic pain and three trials in musculoskeletal conditions. They found that 57% of patients with neuropathic pain achieved at least 50% pain relief with capsaicin, compared with 42% of patient of placebo; for patients with musculoskeletal conditions, the capsaicin response was 38 vs. 25% for placebo (Mason et al. 2004a). Approximately one-third of the patients experienced local adverse events with capsaicin.

Derry et al. (2009) performed a Cochrane review that included six studies (389 participants in total) which compared regular application of low-dose (0.075%) capsaicin cream with placebo cream; the NNT for any pain relief over 6–8 weeks was 6.6 (4.1–17). Two studies (709 participants in total) compared a single application of high-dose (8%) capsaicin patch with placebo patch; the NNT for ≥30% pain relief over 12 weeks was 12 (6.4–70). Local skin reactions were more common with capsaicin, usually tolerable, and attenuated with time; the NNH for repeated low-dose application was 2.5 (2.1–3.1) (Derry et al. 2009).


Topical NSAIDs


Massey et al. (2010) performed a Cochrane Review of topical NSAIDs for acute pain in adults. Forty-seven studies were included; most compared topical NSAIDs in the form of a gel, spray, or cream with a similar placebo, with 3,455 participants in the overall analysis of efficacy. For all topical NSAIDs combined, compared with placebo, the number needed to treat to benefit (NNT) for clinical success, equivalent to 50% pain relief, was 4.5 (3.9–5.3) for treatment periods of 6–14 days. Topical diclofenac, ibuprofen, ketoprofen, and piroxicam were of similar efficacy (Massey et al. 2010).


Topical Salicylate


Topical salicylate has proven superior to placebo in both acute pain (three trials, 182 patients; NNT is equal to 2.1; 95% confidence CI; 1.7–2.8) and chronic pain (six trials, 429 patients; NNT equals 5.3; 95% CI; 3.6–10.2) (Mason et al. 2004b). However, larger more rigorous trials tended to be negative.


Topical Diclofenac


Diclofenac is available in the United States in a topical formulation as: a topical diclofenac sodium 1% (DSG) gel (Voltaren Gel®), a 1.5% diclofenac solution (Pennsaid®), or as a diclofenac epolamine topical patch (DETP) 1.3% (Flector Patch®). Baraf et al. (2011) analyzed pooled data from three randomized, double-blind, parallel-group, placebo-controlled, multicenter trials which evaluated the safety and efficacy of topical diclofenac sodium gel for knee osteoarthritis in elderly and younger patients. They found that diclofenac sodium gel was effective and generally well tolerated in adults regardless of age. These data support the topical application of DSG for relief of OA knee pain in elderly and younger patients (Baraf et al. 2011).

Towheed (2006) performed a systematic review and meta-analysis of randomized controlled trials of Pennsaid® therapy for osteoarthritis of the knee. Pennsaid is an effective topical NSAID in patients with OA of the knee. Apart from minor localized skin reactions, Pennsaid was as safe as VCP (Towheed 2006). Galer et al. (2000) reported the results of a multicenter controlled clinical trial evaluating the topical diclofenac patch for minor sports injury pain. They found that the diclofenac epolamine patch is an effective and safe pain reliever for treatment of minor sports injury pain.

Kuehl (2010) analyzed eight studies utilizing the DETP (1.3%) for patients with acute pain due to soft tissue injuries. The search identified six placebo-controlled clinical studies, one active-comparator-controlled clinical study, and one open-label comparator clinical study of the efficacy and tolerability of the DETP in patients with soft tissue injuries (Kuehl 2010). Three studies reported on tolerability. Primary analyses among the eight studies reported DETP-associated reductions in spontaneous pain from baseline, assessed using a visual analog scale, ranging from 26 to 88% on day 7 and 56–61% on day 14. The use of the DETP was associated with significantly greater reductions in pain scores compared with a placebo patch (two studies) on day 7 (88 vs. 74%; p  =  0.001) and day 14 (56.5 vs. 46.8%; p  =  0.001) and compared with diclofenac diethylammonium topical gel (one study) on day 14 (60.8 vs. 40.8%; p  <  0.001) (Kuehl 2010).


High Concentration Capsaicin Patch


A high concentration (8%) capsaicin patch has been studied in multiple RCTs in patients with postherpetic neuralgia (Backonja et al. 2008, 2010; Irving et al. 2011) and reviewed (Jones et al. 2011). A single high-concentration capsaicin patch application utilized to provide analgesia for patients with painful HIV neuropathy resulted in a mean pain reduction of 22.8% during weeks 2–12 as compared to a 10.7% reduction for controls (p  =  0.0026, Simpson et al. 2008). One-third of high-concentration capsaicin patch-treated patients reported ≥30% pain decrease from baseline as ­compared to 18% of controls (p  =  0.0092). Self-limited, mild-to-moderate local skin reactions were commonly observed (Simpson et al. 2008). However, in a second HIV neuropathy trial, the effects of high concentration capsaicin patches did not meet the primary endpoint of the study (Noto et al. 2009).

Application of the high-concentration capsaicin patch in patients with postherpetic neuralgia appears to be safe and well tolerated, and adverse effects were limited to transient increases in pain, as well as location reactions associated with patch application (e.g., application site reactions [pain, erythema]). After a single 1-h application (Webster et al. 2010), the high concentration capsaicin patch may be associated with some sustained reductions in pain that persist for 2–3 months. Simpson et al. (2010) concluded that repeated treatments administered over a 1-year period are generally safe and well tolerated.


Botulinum Toxin


Botulinum toxin (BTX) is a potent neurotoxin produced by the anaerobic bacterium, Clostridium botulinum. Out of the seven known serotypes, three type-A preparations, BOTOX® (onabotulinumtoxinA, product of Allergan, Inc. Irvine, CA), Xeomin® (incobotulinumtoxinA, product of Merz Pharmaceuticals, LLC, Greensboro, NC) and Dysport® (abobotulinumtoxinA, product of Medicis Pharmaceutical Inc., Scottsdale, AZ) have been developed. Currently, Type B is commercially available as Myobloc® in the United States.

Botulinum toxin Type A (BTX-A) is FDA-approved for multiple medical conditions including the treatment of cervical dystonia and more recently for chronic migraine headaches. It also appears that BTX-A may provide analgesic effect for patients with neuropathic pain (Ranoux et al. 2008).

Xiao et al. investigated the therapeutic benefits of BTX-A in subjects with PHN in a randomized, double-blind, placebo-controlled study. Sixty subjects with PHN were randomly and evenly distributed into BTX-A, lidocaine, and placebo groups (Xiao et al. 2010). Compared with pretreatment, VAS pain scores decreased at day 7 and 3 months posttreatment in all three groups (p  <  0.01). However, the VAS pain scores of the BTX-A group decreased more significantly compared with lidocaine and placebo groups at day 7 and 3 months posttreatment (p  <  0.01). Sleep time (hours) had improved at day 7 and at 3 months compared with pretreatment in all three groups, but the BTX-A group improved more significantly compared with lidocaine and placebo groups (p  <  0.01). The percent of subjects using opioids posttreatment in the BTX-A group was the lowest (21.1%) compared with the lidocaine (52.6%) and placebo (66.7%) groups (p  <  0.01) (Xiao et al. 2010). Thus, subcutaneous administration of BTX-A significantly decreased pain in PHN and reduced opioid use compared with lidocaine and placebo at day 7 and 3 months posttreatment. It also increased subjects’ sleep times (Xiao et al. 2010).


Spinal Analgesics


Opioids have been and continue to be a mainstay agent for intraspinal therapy. The guidelines developed at the Polyanalgesic Consensus Conference suggest that the first-line intraspinal agent should be an opioid alone (e.g., preservative-free, sterile morphine sulfate or hydromorphone) or ziconotide, and suggest switching from one agent to another opioid agent, or adding agents if the suggested “maximum” dose is reached (e.g., 15 mg/day of morphine), if acceptable analgesia is not achieved, or if side effects occur (Deer et al. 2007).


Treatment Considerations for Neuropathic Pain


The International Association for the study of Pain Neuropathic Pain Special Interest Guidelines for the treatment of neuropathic pain recommended three classes of medications as first-line treatments: antidepressants with both norepinephrine and serotonin reuptake inhibition (TCAs and selective serotonin and norepinephrine reuptake inhibitors [SSNRIs]), calcium channel α2-δ ligands (gabapentin and pregabalin) and topical lidocaine (lidocaine patch 5%) (Dworkin et al. 2007). Opioids and tramadol were recommended as generally second-line treatments, except in certain specific situations in which it was recommended that first-line use could be considered. A number of medications were considered as third line treatments.

The guidelines acknowledge that a combination of medication with efficacy for neuropathic pain may provide greater analgesia than use of individual medications as monotherapy, although such combination therapy may also be associated with increased side effects, inconvenience, risk of drug interaction and cost (Dworkin et al. 2007). Nevertheless, because ≤50% of patients in neuropathic pain trials typically achieve satisfactory pain control, many patients in clinical practice will require treatments with combinations of medications. Such combination therapy is incorporated into a stepwise management strategy for patients with partial response to treatment with first-line medications.


Treatment Considerations for Fibromyalgia


Five types of medications are effective in FM (see also Velly et al. 2011) V: (1) TCAs, (2) Cyclobenzaprine, (3) tramadol, (4) SNRIs (duloxetine, milnacipran), and (5) calcium channel α2-δ-ligands (gabapentin and pregabalin). Although classified as a muscle relaxant, cyclobenzaprine has a chemical structure closely related to TCAs, which may partially account for its effectiveness in FM. While trials have shown the efficacy of tramadol in FM, the few studies of potent full mu agonist opioids have not shown benefit. There is no RCT evidence that NSAIDs are effective monotherapy for FM.

A meta-analysis of antidepressants published in 2000 found 13 trials with evaluable data involving three classes of antidepressants: TCAs (9 trials), SSRIs (3 trials), and s-adenosylmethionine (2 trials) (O’Malley et al. 2000). Overall, antidepressants were superior to placebo with an NNT of 4. The effect sizes for pain, fatigue, sleep and overall well-being were all moderate (ES, 0.39–0.52). In the five studies where there was adequate assessment for treatment response independent of depression, only one study found a correlation between symptom improvement and depression scores. Antidepressant class did not make a difference, although only three trials tested SSRIs (O’Malley et al. 2000).

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Oct 16, 2016 | Posted by in PAIN MEDICINE | Comments Off on Evidence-Based Pharmacotherapy of Chronic Pain

Full access? Get Clinical Tree

Get Clinical Tree app for offline access