Pharmacogenetics of Chronic Pain Management Julie L.V. Shaw, Prateek K. Lala, Bhushan M. Kapur PII: DOI: Reference:

S0009-9120(14)00389-0 doi: 10.1016/j.clinbiochem.2014.05.065 CLB 8771

To appear in:

Clinical Biochemistry

Received date: Revised date: Accepted date:

8 March 2014 25 May 2014 27 May 2014

Please cite this article as: Shaw Julie L.V., Lala Prateek K., Kapur Bhushan M., Pharmacogenetics of Chronic Pain Management, Clinical Biochemistry (2014), doi: 10.1016/j.clinbiochem.2014.05.065

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Pharmacogenetics of Chronic Pain Management Julie L.V. Shaw1, Prateek K. Lala2 and Bhushan M. Kapur1,2,3 *

PT

1

TE

Address for Correspondence:

D

MA

NU

SC

RI

Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto. 2 Division of Clinical Pharmacology and Toxicology, The Hospital for Sick Children University of Toronto and 3 Department of Clinical Pathology, Sunnybrook Health Sciences Center.

AC CE P

*Dr. Bhushan M. Kapur Department of Clinical Pathology Sunnybrook Health Science Centre 2075 Bayview Ave Toronto, Ontario M4N 3M5 Tele: (416) 813 8130 Fax: (905) 849 4389

Email: [email protected]

Julie L.V. Shaw current address: Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Department of Pathology and Laboratory Medicine, The University of Ottawa

1

ACCEPTED MANUSCRIPT Pharmacogenetics of Chronic Pain management

PT

Abstract

a) Objective: The experience of chronic pain is one of the commonest reasons individuals seek

RI

medical attention, making the management of chronic pain a major issue in clinical practice. Drug metabolism and responses are affected by many factors, with genetic variations

SC

offering only a partial explanation of an individual’s response. There is a paucity of

NU

evidence for the benefits of pharmacogenetic testing in the context of pain management.

b) Design and Methods: We reviewed the literature between 2000 and 2013, and references

MA

cited therein, using various keywords related to pain management, pharmacology and pharmacogenetics.

D

c) Results: Opioids continue to be the mainstay of chronic pain management. Several non-

TE

opioid based therapies, such as treatment with cannabinoids, gene therapy and epigeneticbased approaches are now available for these patients. Adjuvant therapies with

AC CE P

antidepressants, benzodiazepines or anticonvulsants can also be useful in managing pain. Currently, laboratory monitoring of pain management patients, if performed, is largely through urine drug measurements. d) Conclusions: Drug half-life calculations can be used as functional markers of the cumulative effect of pharmacogenetics and drug-drug interactions. Assessment of half-life and therapeutic effects may be more useful than genetic testing in preventing adverse drug reactions to pain medications, while ensuring effective analgesia. Definitive, mass spectrometry-based methods, capable of measuring parent drug and metabolite levels, are the most useful assays for this purpose. Urine drug measurements do not necessarily correlate with serum drug concentrations or therapeutic effects. Therefore, they are limited in their use in monitoring efficacy and toxicity.

Keywords: chronic pain, pain management, pharmacokinetics, pharmacogenetics, opioids, NSAIDs, triptans

2

ACCEPTED MANUSCRIPT Introduction According to the International Association for the Study of Pain (IASP), pain is defined as an “unpleasant

PT

sensory and emotional experience associated with actual or potential tissue damage, or described in terms of such damage”. Several different subtypes of pain can be described, based on their neuropsychological

RI

basis and duration, including neuropathic, nociceptive, psychogenic, referred, phantom, acute and chronic [1]. Chronic pain is very common, with one in three Americans and one in five Canadians reported to suffer

SC

from this problem[2, 3]. It is one of the most frequent reasons for individuals to seek medical care. If untreated, chronic pain can lead to physical and social dysfunction and diminished quality of life.

NU

Successful pain management provides adequate analgesia without excessive adverse effects. Current pain management strategies largely employ the use of the World Health Organization (WHO) pain ladder,

MA

beginning with non-opioid medications, such as NSAIDs, progressing to weak opioids, and culminating with strong opioids, particularly in cancer pain[4]. The WHO also recommends adjuvant therapy with

D

antidepressant medications to aid in reducing anxiety often associated with chronic pain[4].

TE

Management of pain can be complicated by lack of adherence, the potential for abuse or dependence on the medications used and adverse drug side effects. Many factors can influence drug disposition, including

AC CE P

genetic variation, which can further complicate management of these patients. Genetic studies have identified several loci in which polymorphic changes can influence the pharmacodynamics and kinetics of analgesic drugs. Current patient monitoring in pain management (if performed at all) is largely based on urine drug measurements. However, therapeutic drug monitoring (TDM) via plasma drug levels and half-life may prove more useful in monitoring patients prescribed pain medications to ensure efficacy while minimizing adverse drug reactions. TDM can also be very useful for the identification of drug-related side effects and patient-reported lack of effect (e.g., tolerance) Finally, non-opioid analgesics are often tried as options for management of pain, particularly in individuals where opioids are not a suitable choice. In addition, novel therapies, including targeting of epigenetic changes and gene therapy-based approaches are further broadening future options for the treatment of chronic pain.

3

ACCEPTED MANUSCRIPT

PT

Methods

We conducted an online systematic search for papers and related abstracts published between 2000 and

RI

2013 using the National Library of Medicine database, PubMed. Using the following headings: “pain

SC

management medications”, “pain management drugs”, “chronic pain management”, “pharmacogenetics and pain management”, and “drug monitoring and pain management”, we identified 6670 papers. The abstracts of all papers were reviewed and those that appeared to be relevant to the subject were selected

NU

for further study. References cited therein, including original publications, were also selected for further

MA

review.

D

Pain

Pain is the most common presenting physical symptom in primary care, accounting for an enormous

TE

burden of patient suffering, quality of life, work and social disability, and health care and societal costs[5]. Pain is defined as an unpleasant sensory and emotional experience associated with actual or potential

AC CE P

tissue damage [1]. However, the experience of pain is highly subjective and idiosyncratic, and individuals develop pain thresholds through experiences of injury or pathology in early life [1]. Neuropathic pain results from actual damage to nerve fibers themselves in the central or peripheral nervous system [6]. This damage can lead to nerve dysfunction, causing numbness, weakness and/or loss of reflexes. Common descriptions accompanying neuropathic pain include “burning”, “shooting” or “shocklike” sensations [7]. The most common causes of chronic neuropathic pain include diabetic neuropathy, post-herpetic neuralgia, fibromyalgia, lower back pain (radiculopathy due to disc herniation) and osteoarthritis [8]. Nociceptive pain is caused by stimulation of specialized sensory neurons, called nociceptors, by noxious stimuli including extremes of temperature, or mechanical or chemical excitation. It is often categorized as somatic (derived from skin and deep tissue) or visceral (originating from internal organs). Due to high concentrations of nociceptors in somatic tissues, chronic somatic pain is typically localized, and often results from degenerative or inflammatory processes. Nociceptive pain is frequently described as stabbing, pricking, burning, throbbing or cramping [9].

4

ACCEPTED MANUSCRIPT Psychogenic pain refers to painful sensations caused or amplified by mental or emotional factors with no evident physical tissue damage [10]. Referred pain is perceived at a site adjacent to or distant from the site of painful stimulus; the mechanism for this process is not yet well understood [11]. Phantom pain, a sub-

PT

type of referred pain, is the feeling of painful sensations from a part of the body which has been lost, and from which the brain no longer receives signals. Individuals who have undergone limb amputation often

RI

experience phantom pain [12].

SC

Acute and chronic are terms commonly used to describe the duration of painful sensations, but are difficult to categorically define. In general, acute pain resolves within weeks of the initial causative insult, whereas

MA

NU

chronic pain is continuous, long-term pain which can last months or years [13].

Chronic Pain

D

The IASP defines chronic pain as that pain which persists past the normal time of healing following an injury. However, since determining the end of the healing phase is problematic, most clinical definitions

TE

use a fixed time of persistent pain after initial onset. This demarcation between acute and chronic pain is somewhat arbitrary, and typically ranges from three months (e.g., for post-herpetic neuralgia) to six

AC CE P

months (e.g., for chronic low back pain).[14, 15]. The treatment of chronic pain can be difficult because it is a complex condition influenced by genetic makeup as well as physiological and psychological factors. Chronic pain is a problem in many societies, with prevalence ranging from 2-40% in the adult population, resulting in social and economic impacts [16-18]. Chronic pain can interfere with an individual’s activities of daily living, leading to work or school absenteeism, inability to participate in leisure activities, sleep disturbances and problems eating. In the United States, one in three Americans experience chronic pain and chronic pain is the most common reason for people to visit their family doctor [3]; one in five Canadians are reported to suffer from chronic pain [2]. It is therefore important that chronic pain be diagnosed accurately and treated effectively. Recently, an increased focus on pain management has led to a six-fold increase in the sales of prescription opioids in the U.S. between 1997 and 2006 [19]. Use of analgesics has increased from 7.2% of the U.S. population in 1988-1994 to 10.2% during the period 20072010[20].

5

ACCEPTED MANUSCRIPT Pharmacogenetics and Pain Management Pharmacogenetics refers to the way in which genetic differences between individuals influence patient

PT

drug responses and drug disposition [21]. Empirically, it is well understood that large inter-individual variations exist with respect to the response to analgesics [22].With conventional drug dosing, some

RI

patients will experience toxicity whereas other patients will not receive adequate analgesia from the same dose. Variations in drug efficacy can vary as much as 2- to 10-fold or even 100-fold among members of the

SC

same family [23-25]. These differences can be due to pharmacodynamics factors, based on variations in drug target receptors and downstream signal transduction, and pharmacokinetic factors, which affect drug

NU

metabolism and/or elimination, altering the relationship between drug dose and steady state serum drug concentrations. Development of tolerance, which may occur through both dynamic and kinetic

MA

mechanisms, can also play a significant role in this response variation. Genetic variability is one factor playing a role in the way in which drugs affect physiology. Generally, genes

D

affecting outcome of treatment can be divided into two broad categories: those genes affecting pharmacokinetics, and those affecting pharmacodynamics [22]. In the case of pain management drugs,

TE

genes associated with altered pharmacokinetics include those which encode members of the cytochrome P450 family of enzymes, enzymes responsible for glucuronidation and drug transporter proteins [21, 22].

AC CE P

Genes encoding cyclooxygenase enzymes, the opioid receptors and the enzyme catecholamine methyltransferase (COMT) can affect drug pharmacodynamics [21, 22]. Světlík and colleagues provide an up-to-date review of results from pharmacogenetic studies of candidate genes in both the kinetic and dynamic domains[26]

Cytochrome P450 Enzymes The cytochrome P450 (CYP) enzymes comprise a category of proteins whose biosynthesis is controlled by a large super-family of genes. Members of the CYP family play important roles in drug metabolism and therefore influence the concentration of a drug present in circulation. The primary catalytic function of CYP enzymes is identified as the transfer of one oxygen atom from molecular oxygen into various substrates. Additionally, some CYP enzymes act as isomerases, reductases, dehydrases, or nitric oxide (NO) synthases, and some can catalyse oxidative cleavage of esters [27].

6

ACCEPTED MANUSCRIPT CYPs are responsible for approximately 80% of all phase I metabolism reactions (Figure 1) [27, 28]. Genetic changes in CYP genes can result in alterations in enzyme activities. One CYP isoform which has been extensively studied with respect espect to genetic variation is CYP2D6. This enzyme accounts for a very small

PT

percentage (< 2%) (Figure 1b)) of all CYPs expressed in the human body, but is responsible for the metabolism of >20% of all drugs (Figure 1a) 1a),, including many drugs used in management of pain [29]. Most

RI

of these enzymes are polymorphic [30] [30]; more than 80 CYP2D6 variants have been identified which,

SC

functionally, are associated with four general phenotypes: extensive metabolizer (EM), intermediate metabolizer (IM), poor metabolizer er (PM) and ultra ultra-rapid metabolizer (UM)[31].. The prevalence of these phenotypes vary by ethnicity [22] and there are several examples in which phenotypic differences in

MA

NU

individuals have led to adverse drug reaction reactions.

AC CE P

TE

D

Figure igure 1: Cytochrome p450: Abundance and proportions

a) Proportions of Drugs Metabolized by CYP 450 Enzymes

b) CYP Abundance in human liver

Adapted from: Rendic S and Di Carlo FJ FJ[27]

Adapted from Yeo et. al. [32]

7

ACCEPTED MANUSCRIPT

In 2004, a case reported by Gasche and colleagues described a 62-year-old male, with a history of chronic lymphocytic leukemia, who presented with fatigue, dyspnea, fever and cough with a bronchoalveolar

PT

lavage culture revealing yeast [33]. He was treated with codeine as a cough suppressant. However on day 4 of treatment, he deteriorated rapidly and became unresponsive. He was given naloxone, an opioid

RI

receptor antagonist, which resulted in rapid improvement in his condition. Several factors were at play in

SC

this case, leading to the patient’s rapid deterioration. Genetic studies revealed that he had CYP2D6 duplications, causing him to be an ultra-rapid metabolizer of the pro-drug (codeine) to the active

NU

metabolite (morphine). Furthermore, CYP3A4 is also involved in codeine metabolism, specifically in the conversion of codeine to (relatively inactive) norcodeine. In this case, co-treatment with clarithromycin

MA

and voriconazole, both CYP3A4 inhibitors, resulted in decreased conversion of codeine to norcodeine, shunting more codeine into the CYP2D6 morphine pathway. The patient also developed acute renal failure which resulted in the accumulation of glucuronide metabolites with opioid activity (morphine-6-

D

glucuronide) [33].

TE

Opioids, including codeine, are among the pain medications metabolized by CYP2D6. In 2006, Koren and colleagues reported a case of a breast-fed infant who died at 13 days of age [34]. The infant’s mother had

AC CE P

been prescribed codeine for episiotomy pain. Stored breast milk was found to contain higher than expected levels of morphine (87 ng/mL; expected 1.9-20.5 ng/mL). CYP2D6 genotyping revealed that the infant’s mother was heterozygous for a CYP2D6 duplication, making her an UM of codeine to morphine. The infant died from CNS depression and this, along with the genetic results, were consistent with opioid toxicity resulting from increased conversion of codeine to morphine. This case prompted the FDA in the USA as well as Health Canada to advise physicians against the prescription of codeine to nursing mothers [35, 36]. Studies have also shown that CYP polymorphisms have an influence on the metabolism of non-steroidal anti-inflammatory drugs (NSAIDs) and have the potential to cause adverse drug reactions, most notably gastrointestinal (GI) bleeding, a frequent reaction resulting in significant patient morbidity and mortality. The risk of GI bleeding is particularly high with higher doses of NSAIDs [37]. Many NSAIDs are metabolized by CYP2C9, which has been shown to have two allelic variants, referred to as CYP2C9*2 and CYP2C9*3 [37]. The CYP2C9 enzymes encoded by the *2 and *3 alleles are reported to have 50% and 15% of the activity of the wild-type enzyme, respectively, resulting in poor metabolism, and thus prolonged action, of NSAIDs in individuals with either of these two alleles [38, 39].

8

ACCEPTED MANUSCRIPT Due to their platelet-inhibiting action, NSAIDs are also responsible for an increased risk of bleeding, particularly in patients taking the anticoagulant warfarin [38], since CYP2C9 is also important for warfarin metabolism. Patients with the CYP2C9 *2 or *3 genotypes taking both an NSAID and warfarin have been

PT

shown to have a significantly higher risk for an elevated prothrombin time, compared to patients with the

RI

wild-type 2C9 genotype [38].

Celecoxib is a selective COX-2 inhibitor which is metabolized by CYP2C9. In a study of patients prescribed

SC

celecoxib for rheumatoid or osteoarthritis, patients with the *2 or *3 genotypes showed an increased elimination half-life compared to patients with the wild-type genotype [40]. In a similar study, the

NU

investigators looked at patients prescribed one of several different NSAIDs, including celecoxib, ibuprofen and naproxen. They grouped the patients into those who experienced GI bleeding and those who did not,

MA

and performed CYP 2C9 genotype analysis on all patients. Within the group of patients with GI bleeding, a higher number of individuals had the *2 or *3 genotype compared to the group not experiencing gastrointestinal bleeding [41]. The increased risk of bleeding was believed to result from reduced CYP2C9

AC CE P

P-glycoprotein

TE

D

enzyme activity in patients with the *2 or *3 alleles, leading to increased NSAID concentrations [41].

The ABCB1/MDR1 gene encodes P-glycoprotein, an efflux transporter that influences drug transport in several tissues, particularly the intestine, kidney and brain [42, 43]. P-glycoprotein is highly expressed in endothelial cells of the brain vasculature, and is believed to effect the efflux of drugs across the blood brain barrier [43]. ABCB1/MDR1 is a highly polymorphic gene, with 38 single nucleotide polymorphisms (SNPs) identified in the coding region [44]. The activity of P-glycoprotein is thought to be affected by genetic variability, with certain SNPs leading to decreased P-glycoprotein expression and activity. One study, which examined individuals prescribed oxycodone, found that people homozygous or heterozygous for variant ABCB1/MDR1 alleles reported larger decreases in pain, but also a higher frequency of adverse drug reactions to oxycodone, presumably due to decreased efflux and therefore higher plasma concentrations of the drug [42]. Genetic variability of ABCB1/MDR1 has also been found to be associated with inter-individual differences in pain relief achieved by morphine [43]. In a study of Korean patients receiving intravenous fentanyl, certain ABCB1/MDR1 genotypes were associated with prolonged fentanyl-induced suppression of respiration rate [45].

9

ACCEPTED MANUSCRIPT Cyclooxygenases 1 and 2 Cyclooxygenase 1 and 2 are encoded by two separate genes, PTGS1 (COX1) and PTGS2 (COX2). Genetic

PT

variation in either of these enzymes would be expected to cause altered pharmacodynamic responses to NSAIDs. In a study which investigated post-surgical expression of PTGS1 and PTGS2, in relation to SNPs in

RI

each gene, certain SNPs were shown to be associated with altered expression of the PTGS2 transcript [46]. The authors also reported differences in patient response to two NSAIDs, rofecoxib, a selective COX-2

SC

inhibitor and ibuprofen, a non-selective COX inhibitor. In particular, the G→C polymorphism at -765, in the promoter region of PTGS2, was found to be associated with lower PTGS2 expression in individuals

NU

heterozygous (G/C) or homozygous (C/C) for the minor allele, compared to those homozygous for the major allele (G/G) [46]. Subjects with G/G genotype also reported lower pain intensity with rofecoxib

MA

treatment for 48 hours following surgery, compared to G/C or C/C subjects. This difference was not demonstrated for patients treated with ibuprofen. However, subjects with G/C and C/C genotypes reported decreased pain intensity with ibuprofen treatment at 48 hours following surgery, compared to

D

G/G patients. This difference was not seen in subjects treated with rofecoxib [46]. These data suggest

TE

that patients with increased PTGS2 expression (G/G genotype) benefitted more from treatment with rofecoxib, whereas patients with decreased PTGS2 expression (G/C or C/C genotypes) benefited more from

AC CE P

treatment with ibuprofen [46]. These findings are likely explained by the selectivity of rofecoxib for COX-2, encoded by PTGS2.

Opioid Receptor µ

Opioid receptor µ is the primary site of action for many endogenous opioids, as well as those used for analgesia. A number of SNPs have been described in OPRM1, the gene encoding this receptor. The most well characterized SNP in OPRM1 is A118G. A study by Janicki et al.[47] found the minor allele (G) present at lower frequency in subjects with chronic pain treated with opioids, compared to a control group of opioid naïve patients, suggesting that the G allele may confer protection against pain. Epidural opioids are commonly used to provide analgesia to women during labour. Women heterozygous or homozygous for the OPRM1 A118G allele have been demonstrated to have higher pressure pain thresholds than women homozygous for the more common A allele [48]. The A→G nucleotide substitution leads to an amino acid change from asparagine to aspartic acid, and is thought to result in higher binding affinity of β-endorphin to the opioid-µ receptor. In another study of women in labor, the median effective

10

ACCEPTED MANUSCRIPT dose (ED50) of intrathecal fentanyl required to achieve effective analgesia was significantly higher in women homozygous for the A allele (26.8 µg) compared to women heterozygous or homozygous for the G allele

PT

(17.7 µg) [49].

RI

Catechol-O-methyltransferase (COMT)

SC

The COMT enzyme is responsible for the inactivation of catecholamines such as dopamine, norepinephrine and epinephrine. Several polymorphisms have been identified in the COMT gene; however, the most

NU

widely studied variant is a G to A nucleotide substitution resulting in an amino acid change from valine to methionine at codon 158 (Val158Met). This amino acid change is reported to decrease the thermal

MA

stability of the COMT protein, resulting in reduced enzymatic activity. In studies of cancer patients experiencing chronic pain, patients homozygous for the more common Val 158 were found to require higher doses of morphine to achieve analgesia, compared to patients heterozygous or homozygous for Met

D

158 [50, 51]. These findings suggest that variable COMT activity may influence the effects of opioids.

TE

Animal studies have demonstrated reduced neuronal enkephalin content in the brain with chronic activation of dopaminergic transmission. The reduced enkephalin content has been shown to be followed

AC CE P

by an upregulation of the opioid-µ receptor [52, 53]. The lower doses of morphine required by individuals with the Met158 variant may be explained by reduced COMT activity which is associated with increased dopaminergic stimulation, resulting in upregulation of opioid-µ receptor expression in the brain, making morphine more effective. Another study demonstrated differences in morphine side effects, such as drowsiness, confusion and hallucinations, associated with certain COMT variants, which influenced how well patients tolerated morphine [54].

Clinical use of pharmacogenetics in pain management Ideally, pharmacogenetic studies aim to aid in the selection and dosing of an optimal drug therapy for a specific patient. Choosing the optimal therapy should lead to maximized therapeutic benefit, improved patient adherence and a reduction in adverse drug reactions [55]. The cases of opioid overdose discussed above illustrate instances where knowledge of patient genotypes may have been useful to improve patient outcomes. However, most pharmacogenetic studies to date have examined variability in single candidate genes, such as CYP2D6, and associated outcomes [22]. There are limitations to this approach, as very few

11

ACCEPTED MANUSCRIPT drugs are metabolized by a single enzyme. Genome-wide association studies (GWAS) may possibly improve upon this limitation. Furthermore, in addition to genetics, many other factors affect drug responses, such as patient age, disease co-morbidities and, in particular, co-medication [22, 56] (Figure 2).

PT

Genetics can only partially explain the variability in patient responses to analgesic drugs. Patients are very commonly prescribed several medications for multiple co-morbidities. Some medications induce CYP

RI

enzyme activity [57], as illustrated in the case above [33], while others inhibit activity [57] , which can lead

SC

to altered metabolism unrelated to the patient’s CYP phenotype. In general, pharmacogenetic studies thus far in pain management have failed to yield evidence of improved clinical outcomes associated with knowledge of patient genotypes when prescribing pain medications. As GWAS studies continue, and panels

NU

of gene testing become more widely accessible, pharmacogenetics of pain management may yet become

MA

more clinically useful[58].

AC CE P

TE

D

Figure 2 Genetic and non-genetic factors effecting drug metabolism

Adapted from Stamer et. al.[22] 12

ACCEPTED MANUSCRIPT

PT

Genetics and Pain Susceptibility An individual’s genetic susceptibility to chronic pain is thought to be complex, involving multiple genes,

RI

similar to the genetics of other chronic diseases, such as diabetes [21]. As discussed above, several

SC

candidate genes have been identified, with specific mutations shown to alter pain perception profiles between individuals. Examples include: OPRM1, the gene encoding the µ-opioid receptor; COMT,

NU

encoding catechol-O-methyltransferase, involved in the metabolism of catecholamines; GCH1, encoding an enzyme involved in phenylalanine metabolism and the production of dopamine; the melanocortin-1 receptor with mutations showing sex-specific differences in pain perception; and members of the

MA

cytochrome P450 enzyme family, important for the biotransformation of many drugs. Genome-wide association studies and other types of genetic investigations will likely identify many more genes involved

AC CE P

Chronic Pain Management

TE

D

in pain perception.

Successful pain management can be viewed as providing adequate analgesia without excessive adverse effects [59]. In 1982, Rane and colleagues [60] proposed a pharmacological approach to treat cancer pain with morphine. They proposed a step-wise approach that was later adopted by the World Health Organization (WHO) in 1986[61, 62]. Although this WHO stepladder approach was originally aimed at the treatment of chronic cancer pain (Figure 3) [61, 62], it is also widely used in the treatment of chronic noncancer pain [63]. The WHO analgesic ladder recommends initial treatment of pain with non-opioids, such as NSAIDs and acetaminophen. If pain persists, treatment with a weak opioid, such as codeine or tramadol is recommended, followed by a strong opioid, such as morphine, until the patient is free of pain. At each stage of the treatment ladder, adjuvant medications, such as antidepressants or anticonvulsants, may also be given to aid in alleviating patient anxiety; some of these adjuvant drugs may also act directly to counter pain [4, 61]. Evident from this scheme is the fact that opioids remain the mainstay of chronic pain management and will be discussed further below.

13

ACCEPTED MANUSCRIPT

MA

NU

SC

RI

PT

Figure 3 The WHO three-step analgesic ladder

D

Adapted from [60, 61]

AC CE P

TE

Legend: The pain ladder shows a progressive rationale for stronger opioids. Before a new medication is introduced, it is important to wait until the previously administered drug has reached a “steady state” or full effect is achieved. *Adjuvants: antidepressants, anticonvulsants, steroids

Drugs used in pain management

NSAIDs and Acetaminophen in Pain Management (Table 1) NSAIDs such as aspirin and ibuprofen, as well as acetaminophen, act by inhibiting the enzymes cyclooxygenase-1 and -2, which catalyze the synthesis of prostaglandins. The inhibition of prostaglandin synthesis results in the analgesic, anti-pyretic and anti-inflammatory properties of these drugs, with the exception of acetaminophen, which does not show anti-inflammatory affects. Due to the widespread use of NSAIDs, they are the drugs most commonly associated with adverse effects, the most common of which are those involving the GI tract. Reduced prostaglandin levels in the gastrointestinal mucosa decrease mucus and bicarbonate secretions, thereby reducing their protective effects against the acidic gastric

14

ACCEPTED MANUSCRIPT environment. In addition, NSAIDs can be directly toxic to the gastric mucosa, leading to ulceration, and potentially fatal bleeding in the most extreme cases. Furthermore, reduced prostaglandin synthesis in the kidney has been shown to cause renal impairment. Chronic or acutely toxic acetaminophen exposure can

PT

lead to liver toxicity through hepatocellular necrosis [64, 65]. Additionally, aspirin (acetylsalicylic acid) is associated with Reye’s syndrome, primarily affecting children [66, 67], and high doses of salicylates can

RI

cause metabolic acidosis and respiratory depression. More recently, cardiovascular risks associated with

SC

the use of NSAIDs have gained attention. NSAIDs, particularly COX-2 selective agents, such as celecoxib, have been reported to increase the risk for thrombotic events, myocardial infarction and stroke in patients with coronary artery disease and atherosclerosis [68, 69]. These findings have prompted the American

NU

Heart Association to recommend use of acetaminophen or non-acetylated salicylates for the treatment of chronic pain in patients with coronary artery disease [70]. Although classified with nonsteroidal anti-

MA

inflammatory agents, acetaminophen has minimal peripheral anti-inflammatory activity. IV acetaminophen has become a part of the multimodal analgesia approach for pain. Intravenous

D

acetaminophen was made available over 10 years ago in Europe, but in the United States only very

TE

recently. Three randomized placebo controlled trials provided the bases of its approval. Clinical studies show that for management of pain, intravenous acetaminophen injection was at least as effective as

AC CE P

morphine injection in renal colic and oral ibuprofen after cesarean delivery. In children a single-dose acetaminophen injection was similar to meperidine intramuscular (IM) for pain after tonsillectomy.[71]. Effects begin 5 to 10minutes after IV administration, and peak effects occur in about 1 hour with an effective duration of 4 to 6 hours. Table 1 provides pharmacokinetic details of specific NSAIDs and acetaminophen, as used in pain management.

Opioids and Pain Management (Table 2) Opioids remain the most potent analgesics available and are the mainstay of chronic pain management in both cancer patients and patients with non-malignant pain [2, 72]. To manage chronic cancer pain, oral morphine is commonly administered at regular intervals. Chronic dosing can lead to pharmacological tolerance, but not to therapeutic failure. In simple pharmacological terms, opioid-induced tolerance can be described as a “shift to the right” in the dose-response curve; that is, a higher dose is required over time to maintain the same level of analgesia. Different mechanisms have been proposed for the development of tolerance. Säwe and colleagues [73] investigated the metabolic correlate to the development of tolerance to therapeutic effects by measuring morphine, morphine-3- and morphine-6-glucuronide in plasma and

15

ACCEPTED MANUSCRIPT urine samples. They showed that the conjugation of morphine with glucuronic acid is proportional to the dose during long-term treatment with escalating doses, suggesting that this metabolic pathway is neither subject to auto-induction nor saturation. Both in vitro and in vivo studies have shown that, on the cellular

PT

level, chronic opioid treatment leads to a rapid reduction of agonist response, accompanied by internalization of opioid receptors[74]. These early adaptive processes as well as long-term adaptations,

RI

such as receptor downregulation, or counter-regulatory processes such as adenylate cyclase

SC

superactivation, have been suggested to be crucial to the development of opioid tolerance[75-77].

In recent years, there has been an increased focus on pain management which has resulted in a societal

NU

escalation in opioid use, with opioids being currently among the most commonly prescribed medications in developed nations [78]. Despite this, the suitability of opioids for chronic non-cancer pain treatment is

MA

under debate. The efficacy of opioids in this clinical situation has only been reported in short-term trials, and evidence for their overall benefit in long-term therapy is lacking [79].

D

The analgesic properties of opioids begin with their binding to opioid receptors in the central nervous

TE

system (CNS). There are three opioid receptor subtypes, mu (µ), kappa (κ) and delta (δ), which differ in their function and specificity for the drugs they bind. Opioids themselves can be classified, based on their

AC CE P

interactions with these receptors, as agonists, mixed agonists-antagonists and antagonists. The binding of opioids to their G-protein coupled receptors produces signals causing hyperpolarization of neuronal cell membranes and the suppression of neurotransmitter release, resulting in analgesia [78, 80, 81].

Issues with opioids

The increased use of opioids for pain management has led to an expansion in opioid misuse, resulting in an increased number of emergency room visits related to drug-seeking behavior as well as in the number of opioid-related overdose deaths [82, 83]. Although opioids are among the most commonly used analgesics, their clinical use is limited by the development of tolerance and physical dependence. Another growing concern relates to the diversion of opioids from patients to other individuals not under medical supervision. Opioids have several well-known side effects, including nausea, sedation and bowel dysfunction [84]. The most serious potential adverse effect is respiratory depression and failure, which is the most common cause of death related to opioid use [80]. Prolonged use leads to tolerance, resulting in higher dose

16

ACCEPTED MANUSCRIPT requirements, which can lead to physical dependence, demonstrated by withdrawal symptoms in patients experiencing chills, muscle aches, vomiting, diarrhea, hyperthermia, anxiety and hostility [80]. Additionally, opioid use can result in feelings of euphoria which can promote compulsive use leading to psychological

PT

dependence.

RI

A detailed medical assessment is recommended prior to prescription of opioids. This should include a detailed medical history, review of medical records, urine toxicology screen and psychological evaluation,

SC

including questionnaires aimed at determining an individual’s risk for abuse [84, 85]. This is of particular importance in those populations at higher risk for opioid misuse. Chronic pain frequently occurs in

NU

individuals with psychological conditions, including anxiety and depression; it is estimated that 30-60% of individuals with depression also experience chronic pain [86]. Psychological conditions are often comorbid

MA

in individuals with substance-abuse disorders, resulting in a high risk for opioid abuse in many chronic pain patients including those with substance abuse issues and mental health disorders [85].

D

Newer opioid formulations have been designed in efforts to prevent abuse. These include Embeda™, an extended-release morphine co-formulated with the opioid antagonist naltrexone, produced by Pfizer; and

TE

an extended-release form of oxycodone, called OxyNeo™, by Purdue Pharma [84]. OxyNeo™ contains polyethylene oxide, which forms a viscous gel when in contact with water, rendering it unsuitable for

AC CE P

injection or snorting, and also more difficult to chew or crush [87]. Buprenorphine, a semi-synthetic opioid was approved by the FDA in 2002. Sublingual preparations, used in addiction medicine, are co-formulated with the antagonist naloxone to prevent abuse.

Specific Opioids used in Pain Management Opioids can be classified as either weak or strong, depending on their “analgesic ceiling”. This term is defined as the dose above which a drug has no further effect on pain [88]. Weak opioids include tramadol, codeine and hydrocodone; strong opioids include morphine, methadone, buprenorphine, fentanyl, oxycodone, hydromorphone, oxymorphone, meperidine and levorphanol. Detailed pharmacokinetic information for each of these drugs can be found in Table 2. Weak opioids Codeine

17

ACCEPTED MANUSCRIPT Codeine is considered a weak opioid, and is used for its analgesic, anti-tussive and anti-diarrheal properties. It is a pro-drug which requires biotransformation via hepatic CYP2D6 to the active metabolite morphine [89]. Approximately 5-10% of codeine is converted to morphine in the liver, while the remainder is either

PT

conjugated, forming codeine-6-glucuronide (active), or methylated via CYP3A4, producing norcodeine

SC

RI

(inactive) [89].

Hydrocodone

NU

This is a semi-synthetic drug, similar to codeine in structure and to morphine in its effects [89]. Hydrocodone is metabolized by CYP2D6 to active hydromorphone, which binds to the µ-opioid receptor,

MA

and by CYP3A4 to the inactive metabolite norhydrocodone [90].

D

Tramadol

TE

Tramadol is a synthetic codeine analog that binds to the µ-opioid receptor and inhibits the reuptake of

AC CE P

serotonin and norepinephrine [89]. Tramadol has a lower risk of addiction than other opioids; however, it is associated with two significant adverse drug reactions: seizure and serotonin syndrome [91]. Tramadol seizures usually occur in patients already taking anticonvulsant medications. Serotonin syndrome is characterized by neuromuscular and autonomic hyperactivity as well as altered mental status, due to excess serotonin activity in the CNS. Higher risk of this syndrome is associated with tramadol overdose or co-administration of antidepressant medications, such as tricyclic antidepressants and selective serotonin reuptake inhibitors (SSRIs), which, in addition to tramadol, inhibit serotonin reuptake, leading to high serotonin levels in the synaptic space[92, 93]. Tramadol is metabolized by CYP2D6 in the liver to its main metabolite, O-desmethyl-tramadol which shows a higher affinity for opioid receptors than the parent drug [94]. Metabolism to the minor metabolite, Ndesmethyl-tramadol occurs via CYP2B6 and CYP3A4 [95].

Strong opioids Morphine

18

ACCEPTED MANUSCRIPT Morphine increases the threshold for pain perception by binding strongly to the µ-opioid receptor; it also has activity towards κ- and δ-opioid receptors. Morphine administered orally undergoes a large first-pass effect, resulting in oral bioavailability of 38% on average [96]. The major morphine metabolites result from

PT

glucuronidation by the hepatic isoenzyme UGT2B7 to inactive morphine-3-glucuronide (approximately 60%) and active morphine-6-glucuronide (up to 10%)[97]. CYP2D6 plays a minor role in morphine

RI

metabolism. Side effects of morphine include sedation, nausea, a feeling of warmth, urinary retention,

SC

euphoria, reduced ability to concentrate and constipation. The most serious side effect of morphine is potentially fatal respiratory depression with morphine toxicity [89, 98].

NU

Hydromorphone

Hydromorphone, a hydrogenated ketone of morphine, is a synthetic opioid with effects similar to

MA

morphine [89]. When given orally, hydromorphone is rapidly absorbed by the GI tract and undergoes extensive first-pass metabolism in the liver. Like morphine, it is glucuronidated in the liver to

D

hydromorphone-3-glucuronide and hydromorphone-6-glucuronide; hydromorphone-3-glucuronide has no analgesic properties, but has been shown to exhibit neuroexcitatory effects [99]. Hydromorphone

TE

suppresses the cough reflex, similar to other opioids. Side effects of hydromorphone are dose-related and

AC CE P

can include mood changes, euphoria, nausea, vomiting, and hypotension, as well as respiratory depression in large doses [89].

Fentanyl

Fentanyl is a highly lipophilic, synthetic opioid which is 100 times more potent than morphine. Its potency is due to its efficiency in crossing the blood brain barrier, rapidly gaining access to the central nervous system [89, 100, 101]. Given intravenously, fentanyl has a very rapid onset but short duration of action, and is thus administered as a continuous infusion. In managing pain, fentanyl is most often provided through transdermal patches or buccal tablets [89]. This allows the drug to distribute throughout fatty tissues, leading to slower release and prolonged effects. Fentanyl undergoes extensive pulmonary firstpass metabolism as well as hepatic metabolism to the inactive metabolite norfentanyl via CYP3A4 [89]. Remifentanil Remifentanil is a µ-opioid receptor agonist with an analgesic potency similar to that of fentanyl. It was studied for analgesic efficacy correlating with expression of the serotonin transporter (5-HTT), as serotonin

19

ACCEPTED MANUSCRIPT can influence the antinociceptive effects of opioids at the spinal cord. Remifentanil has a significantly better analgesic effect in individuals with a genotype coding for low 5-HTT expression (SA/SA and SA/LG) compared to those with high expression(LA/LA) [102]. It is predominantly metabolized by non-specific

PT

esterases [103], and because of its rapid systemic elimination and ultra-short half-file, it has a pharmacokinetic advantage in clinical situations requiring predictable termination of effect such as

RI

analgesia during labor[104]. In a systematic review, Leong and colleagues compared remifentanil with

SC

meperidine for labor analgesia. They found remifentanil to be superior in reducing mean visual analog scale pain scores for labor pain after 1 hour [105]. Remifentanil crosses the placenta, but is rapidly

MA

without adverse neonatal or maternal effects[106].

NU

metabolized and redistributed. Although maternal sedation and respiratory changes do occur, they are

Methadone

D

Methadone is a synthetic opioid with morphine-like effects. It is commonly used in the treatment of opioid addiction, but also in the treatment of chronic pain [89, 107, 108]. Given orally, methadone has a much

TE

higher bioavailability compared with oral morphine (70-90%, compared to 38%, respectively). Due to its relatively slow metabolism and high lipid solubility, methadone also has a long half-life (between 8 and 60

AC CE P

hours), leading to longer term analgesia than with morphine [107, 108] Methadone is extensively metabolized by CYP3A4, and to a lesser extent by CYP1A2, -2D6, -2D8, -2C9/2C8, -2C19, and -2B6, to its inactive metabolite, 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) [107-110]. Caution must be exercised when co-prescribing methadone with QT-prolonging cardiac drugs due to methadone’s ability to interact with voltage-gated potassium channels in the myocardium, also leading to QT prolongation [111] and ventricular arrhythmias such as Torsades de Pointes. Significant drug-drug interactions have been reported for methadone, when combined, for example, with fluoxetine, quetiapine, or promethazine [110].

Buprenorphine As an alternative to methadone, buprenorphine can be used for the treatment of opioid addiction [89]. It has also been approved for the treatment of moderate to severe pain [112]. Similarly to morphine, buprenorphine must be used with caution in the setting of co-administration of other QT-prolonging medications or in individuals with hyperkalemia [112]. Buprenorphine has a ceiling effect with respect to respiratory depression, thereby reducing the likelihood of this potentially fatal consequence, making it an

20

ACCEPTED MANUSCRIPT attractive choice for analgesia [113]. Given orally, buprenorphine undergoes extensive first-pass metabolism and is therefore usually administered by transdermal or buccal routes to improve bioavailability [112]. Prolonged analgesia can be achieved with buprenorphine due to its lipophilic

PT

properties and its slow dissociation from opioid receptors. Buprenorphine is metabolized by CYP3A4 in the

RI

liver primarily to nor-buprenorphine.

SC

Oxycodone

Oxycodone is a synthetic opioid, similar in structure to codeine and prescribed for moderate to severe pain [89]. This drug is commonly compounded with other drugs, such as acetaminophen or ibuprofen [89]. Side

NU

effects of oxycodone treatment include euphoria, sedation, constipation, cough suppression and respiratory depression. The euphoria caused by oxycodone is quite pronounced and can therefore lead to

MA

abuse of and dependence on the drug. Oxycodone is metabolized, via CYP2D6, to the potent metabolite oxymorphone (see below) as well as noroxycodone, a weakly active metabolite [114].

D

Oxymorphone

TE

Oxymorphone offers excellent analgesia, comparable to that of morphine, but with less sedative effects [115]. This results from the lipophilic nature of oxymorphone, allowing ease of access to the central

AC CE P

nervous system. Oxymorphone undergoes extensive hepatic first-pass metabolism, and its major route of metabolism is via glucuronidation [89, 115]. Meperidine (Pethidine)

Meperidine (Demerol™), also known as pethidine, is a synthetic µ-opioid receptor agonist that is

structurally similar to atropine. Pharmacologically, it represents both morphine and atropine. This unique property of an analgesic with spasmolytic and sedative action accounts for its wide range of clinical applications [116]. Although meperidine efficacy has been questioned [117], a survey of intrapartum analgesia practice in United Kingdom and Norway reported that this opioid is still commonly used during labor. [118, 119].

Levorphanol

21

ACCEPTED MANUSCRIPT Levorphanol is a synthetic opioid, similar in structure to morphine, but more potent in its analgesic effects [89, 120], and also has anticholinergic effects [120]. This drug is usually given orally or intravenously and is

PT

eliminated through glucuronidation in the liver [120].

RI

Other medications in the treatment of chronic pain

SC

Antidepressants

Tricyclic antidepressants (TCAs) and selective serotonin re-uptake inhibitors (SSRIs) are commonly used as

NU

adjuvant therapy in the treatment of chronic pain. The mechanisms by which these medications alleviate pain are not fully understood, but are believed to be mediated through their inhibition of neurotransmitter

MA

reuptake in the synaptic cleft. These mechanisms are related to pain signaling in descending spinal pain pathways [121]. In addition to their analgesic role, antidepressants are commonly used in pain

D

management because depression is often a co-morbidity of chronic pain. Patients experiencing chronic pain have been shown to have 2-5 times the risk of developing depression than the general population

TE

[122].

AC CE P

Common TCAs used in pain management include amitriptyline and imipramine (Table 3), administered at much lower doses for pain management than for psychiatric indications [22]. These two drugs are metabolized to the active metabolites nortriptyline and desipramine, respectively, by hepatic CYP2D6 [22]. Side effects of TCAs that can limit their use include weight gain, anticholinergic effects, hypotension and cardiovascular effects [22, 121]. TCAs have been better characterized for their role in pain management than SSRIs [121].SSRIs used in pain management include fluoxetine and citalopram (Table 3). SSRIs are often better tolerated than TCAs, due to their milder side effects; however, data regarding their efficacy in the treatment of chronic pain have been inconsistent [121]. More recently, antidepressants able to target several neurotransmitters, such as venlafaxine (serotonin, norepinephrine, and dopamine), duloxetine (serotonin and norepinephrine) and bupropion (serotonin and dopamine) have been more widely used in pain management (Table 3). Venlafaxine has been shown to be particularly efficacious when prescribed in combination with gabapentin, a GABA analogue often used to treat neuropathic pain [123]. Anticonvulsants

22

ACCEPTED MANUSCRIPT Anticonvulsants (Table 4) have been used in pain management since the 1960’s when these drugs were originally introduced for the treatment of epileptic seizures [124]. These medications work by a variety of mechanisms to suppress the rapid firing of neurons. Phenytoin, one of the first anticonvulsants introduced,

PT

reduces neuronal excitability by blocking sodium channels. It has been used in the treatment of chronic neuropathic pain, but today its use for this indication is limited due to side effects, namely sedation and

RI

motor disturbances [125]. Carbamazepine has largely replaced phenytoin in chronic pain management

SC

and has been shown to be effective in the treatment of neuropathic pain, in particular trigeminal neuralgia [124, 126] (a disorder characterized by episodes of intense facial pain, originating from the trigeminal nerve) [126]. Side effects of carbamazepine include drowsiness, blurred vision, nausea and vomiting.

NU

There is a dose-effect relationship and it can be difficult to achieve a therapeutic effect in patients who are sensitive to these side effects [125]. This is of particular relevance in elderly patients, where conditions

MA

such as cardiac disease and hypernatremia can further complicate adverse effects [127]; thus, close monitoring of these patients is required [124, 125].

D

Newer anticonvulsants, such as lamotrigine and gabapentin, are showing promise in chronic pain

TE

management; gabapentin, in particular, has been shown to be well tolerated with few side effects [128]. Gabapentin binds to voltage-gated calcium channels, leading to a reduction in the release of the

AC CE P

neurotransmitters glutamate and substance P. It has demonstrated effectiveness in the treatment of diabetic neuropathy, post-herpetic neuralgia, trigeminal neuralgia, multiple sclerosis, migraine as well as in chronic pain caused by malignancy [128]. Lamotrigine has been shown to be useful in the treatment of trigeminal neuralgia and diabetic neuropathy [124]. However, careful titration of lamotrigine dose is required because of the risk of Stevens-Johnson syndrome, a life-threatening dermatological condition leading to cell necrosis, causing the epidermis to separate from the dermis [129].

Triptans Triptans are serotonin agonists with high affinity for the 5HT1B and 5HT1D receptors [130, 131]. The serotonergic system is known to play a role in the pathophysiology of migraines, leading to the development and prescription of triptans for this condition. Binding of these drugs to 5HT receptors in the brainstem and thalamus leads to the blockade of vasoactive peptide release from perivascular trigeminal neurons, and thus reduced cerebral vasodilation [131]. Currently, there are seven triptan molecules available, each with its unique pharmacokinetic and pharmacodynamic profile. The four most commonly

23

ACCEPTED MANUSCRIPT prescribed and well characterized triptans are sumatriptan, naratriptan, zolmitriptan and rizatriptan (Table 5) [132]. Triptans are not recommended for migraine treatment in individuals with poorly controlled hypertension, hepatic or renal impairment or coronary artery disease, as serotonergic blockade may

RI

PT

exacerbate these conditions [131].

SC

Benzodiazepines

Benzodiazepines play a role in the treatment of insomnia, muscle tension, and, in particular, anxiety related

NU

to chronic pain [133]. Chronic pain patients experience increased anxiety compared to the general population, and this can often worsen the subjective experience of pain. Evidence suggests that

MA

benzodiazepines are only effective in treating acute anxiety related to chronic pain, but are not effective in the setting of chronic anxiety [133] (for which antidepressants have been found to be more effective). The caveats to benzodiazepine treatment for chronic pain are the potential for abuse as well as side effects,

AC CE P

Cannabinoids

TE

D

including paradoxical reactions (increased hostility, psychosis and behavioral disturbances) [133].

Cannabis is an annual plant that grows wild in regions of mild or tropical weather, and has been used to treat pain for centuries. It is by far the most widely cultivated, trafficked and abused illicit drug; half of all drug seizures worldwide are of cannabis. The geographical spread of those seizures is also global, covering practically every country of the world. Delta-9-tetrahydrocannabinol (THC) is the active component in cannabis, responsible for its analgesic and psychoactive effects. Ingestion of THC results in feelings of euphoria, relaxation and a general sense of well-being. However, heavy cannabis use can result in hallucinations, anxiety and depression [134, 135]. Endogenous cannabinoids, referred to as endocannabinoids, have been identified, as well as two endocannabinoid receptors, the G-protein coupled receptors CB1 and CB2. High levels of CB1 are expressed in the brain and spinal cord, particularly in areas known to be involved in nociception. CB2 is not as well characterized, but has been shown to have lower expression levels in the brain than CB1. Endocannabinoids are bioactive lipids which regulate neural activity by binding to CB1 and CB2, causing inhibition of neurotransmitter release. Endogenous and exogenous cannabinoid activity results in anti-

24

ACCEPTED MANUSCRIPT nociceptive effects as well as short-term memory impairment, stimulation of appetite and antiemetic effects [134-136].

PT

THC acts synergistically with other analgesic agents, such as morphine and some NSAIDs, though the mechanism for the synergistic action between morphine and THC is unknown [134]. In the case of NSAIDs,

RI

cannabinoids and prostaglandin share a similar structure which is believed to result in a convergence of signals. Activity of CB1 receptors is increased by NSAIDs including ibuprofen and indomethacin.

SC

Furthermore, CB1 receptor antagonists block the analgesic effects of ibuprofen [134]. Evidence for the use of cannabinoids in pain management is lacking, due to the small number of clinical

NU

trials in this area. The best evidence for cannabinoid use in chronic pain comes from HIV and multiple sclerosis studies in which cannabinoids were used to treat the chronic neuropathic pain associated with

MA

these conditions [136]. Smoking cannabis 3-5 times per week was shown to be effective in alleviating neuropathic pain associated with HIV as well as AIDS-related anorexia [137, 138]. Several studies

D

demonstrated benefit of oral treatment with synthetic THC for central pain and spasticity associated with

TE

multiple sclerosis [139-142].

There are several issues associated with the use of cannabinoids in pain management. Firstly, the method

AC CE P

of drug delivery poses a problem. Smoking of cannabis is the easiest delivery route and allows for adequate control of dose [135, 136]. However, the adverse effects of smoking are well known, with THC contents varying depending on where and how the cannabis plants are grown. Canada, which allows prescription of cannabis for chronic pain therapy, requires that patients obtain their medicinal cannabis from a centralized source so that the THC content may be monitored over time [136]. Similar strategies are now under consideration in different parts of the world including the United States. Synthetic cannabinoids, such as dronabinol and nabilone, are available in the U.S. and Canada for use as antiemetics during cancer chemotherapy, and as adjunct therapies for neuropathic and chronic pain. They are not yet widely used, and are under further study to delineate their efficacy and pharmacological parameters in various clinical contexts [143]. Side effects include increased heart rate, blood pressure changes, anxiety, psychomotor retardation, impaired memory and psychosis. Cannabinoids are contraindicated in pregnancy, uncontrolled hypertension, active ischemic heart disease, arrhythmias and schizophrenia. Clark and coworkers suggest guidelines for the use of these cannabinoid compounds [144] Cannabis and cannabinoids have a potential for dependence, which can lead to preoccupation and compulsion. Prescription of cannabinoids for chronic pain management requires clinician to appropriately

25

ACCEPTED MANUSCRIPT select patient and adequately monitor to avoid the development of dependence [134, 136]. Recently, Donoghue and colleagues report the interaction effect of cannabis use on gender and age of onset of schizophrenia and schizoaffective disorder. They show that use of cannabis is associated with an earlier age

RI

gender difference in age of onset is diminished in cannabis users [145].

PT

of onset of schizophrenia. There is a significant interaction between gender and cannabis use whereby the

SC

Ziconotide

Ziconotide is a synthetic conopeptide that selectively inhibits N-type voltage-gated calcium channels, reducing the release of pain-modulating neurotransmitters. Conopeptides are derived from the venom of

NU

predatory cone snails, and ziconotide is specifically derived from the venom of a Pacific fish-hunting snail,

MA

Conus magus [146].

Ziconotide has been approved for intrathecal administration by continuous infusion for the treatment of severe chronic pain in situations where patients are not responding to conventional therapies [147].

D

Evidence indicates that this drug is useful in the treatment of both neuropathic and non-neuropathic pain.

TE

Intrathecal administration is required because, given intravenously, ziconotide demonstrates poor penetration of the blood brain barrier and results in sympatholytic effects leading to orthostatic

AC CE P

hypotension [147]; oral preparations are not available. Ziconotide is highly hydrophilic and is thought to move slowly through the CSF to its target receptor, leading to a lag time to analgesia upon administration. The efficacy of ziconotide shows a wide inter-individual variation [147, 148]. The most common side effects of ziconotide include dizziness, confusion, ataxia, memory impairment and hallucinations. However, there is also large inter-individual variation in the experience of side effects [147]. Toxicity appears to be related to the rate of infusion and is reversible after discontinuation, but, because of low tissue diffusion, the effects can persist for several weeks [147, 148]. Data regarding ziconotide metabolism are sparse. The drug is not believed to be metabolized in the CSF but rather cleared by transport into the systemic circulation where it is degraded by serum proteases [147].

Novel therapies in pain management Targeting epigenetic modifications Epigenetics refers to functionally relevant genomic changes not involving alterations in nucleotide sequence, in response to developmental or environmental cues. Epigenetic mechanisms are manifested

26

ACCEPTED MANUSCRIPT through dynamic, reversible chemical modifications in the genome and are involved in differential gene expression throughout life [149]. The most well characterized epigenetic modifications are DNA

PT

methylation and histone acetylation. DNA methylation is regulated by DNA methyltransferases, while histone modifications are largely regulated

RI

by histone deacetylases (HDAC) [149]. Abnormal DNA methylation [150] and histone acetylation [151, 152] have been demonstrated to occur in pain states. It has also been proposed that primary injuries may

SC

result in lasting epigenetic marks leading to an increased risk of chronic pain [149], which may even persist trans-generationally. Drugs aimed at targeting the enzymes responsible for epigenetic modifications are

NU

under development and in some cases have been approved by the FDA. While these drugs have been developed largely for the purpose of cancer treatment, they are also believed to have potential in pain

MA

management. In animal injury models, rat hind-paw injection of Freund’s complete adjuvant (CFA) followed by intrathecal injection of zebularine, a DNA methyltransferase inhibitor, was shown to reduce pain sensitivity [153]. In a similar rat model, pre-injection treatment with HDAC inhibitors was reported to

D

delay CFA-induced thermal hyperalgesia whereas post-injection treatment was shown to reduce the

TE

intensity of CFA-induced thermal hyperalgesia [154]. In human patients with type 2 diabetes, treatment with valproic acid, which is a known to act as a strong the HDAC inhibitor, conferred improved pain

Gene therapy

AC CE P

sensitivity scores after treatment compared to before treatment [155].

Gene therapy involves the use of viral vectors where the viral genome is replaced with nucleic acid sequences encoding a promoter to drive gene expression as well as a transgene of interest [156]. For pain management therapies, the transgene encodes for an analgesic agent. Gene therapy based approaches have several advantages in pain management. In contrast to pharmacotherapies, gene therapy allows for persistent expression of a protein-based analgesic agent at the site of action. These methods avoid the first-pass effect and can improve bioavailability [156]. Gene therapy can also reduce side effects associated with systemic drug use. There are limitations to these approaches, including inadequate transgene expression and the evocation of immune responses against the viral vectors [156]. Nevertheless, new paradigms using non-viral insertion of therapeutic transgenes are underway to bypass some of these limitations.

27

ACCEPTED MANUSCRIPT In a recent study, terminal cancer patients were given different gene doses of human PENK, encoding preproenkephalin, the protein precursor that is processed to 6-met-enkephalin and 1-leu-enkaphalin, endogenous opioid peptide ligands for the delta opioid receptor [157]. Patients in the two highest virus

PT

dose groups reported decreases in numeric rating scale (NRS) pain scores of up to 50% from before treatment. Patients in the higher dose groups reported substantial reductions in pain compared to

RI

patients in the lowest dose group. This study demonstrates proof-of-concept for gene therapy in pain

NU

Therapeutic Drug Monitoring in Pain Management

SC

management.

MA

In pain management, the rationale for drug monitoring has largely focused on monitoring patient adherence, detecting diversion, and detecting the presence of illicit, non-prescription drugs. Traditionally,

D

drug monitoring in pain management patients has been in the form of urine drug testing. Urine drug testing has been demonstrated to reduce illicit drug use and has been widely used in monitoring

TE

patients in treatment for drug addiction [158, 159]. However, most urine methodologies have been adapted from occupational deterrent-based testing for illicit drug use and have not been optimized for use

AC CE P

in the pain management setting [160]. Given the chronicity of treatment drug administration, and the wide range of drug half-lives (Tables 1-5) a “positive” urine result is to be expected if a patient is adherent in taking his/her medication. A “negative” urine results suggests non-compliance with the treatment protocol or urine sample tampering. To be used effectively in monitoring pain management patients, urine drug testing requires understanding of the principles of drug pharmacokinetics and pharmacodynamics, and an understanding of the type of information urine drug testing can provide along with an appreciation of the limitations of such testing [161]. Urine drug concentrations are often calculated relative to urine creatinine concentrations in order to correct for in vivo dilution, which can vary through time, depending on an individual’s fluid intake. However, these calculations assume stable renal function and creatinine production, an assumption which can lead to errors if unjustified. Pain medications are usually prescribed to be taken in a chronic manner so that steady state plasma concentrations and therapeutic pharmacodynamic effects can be achieved. Steady state drug concentrations in serum imply that the urine excretion window (UEW) of the drug/metabolite is also at “steady state” in patients with stable renal function. Figure 4 shows the concentration of methadone metabolite 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) in a

28

ACCEPTED MANUSCRIPT patient receiving methadone chronically. This figure clearly shows that urine creatinine correction does not add value to the EDDP adherence monitoring. Here, patient non-adherence can be detected when the urine EDDP concentration falls outside the UEW [162]. Extremely low creatinine concentrations are

PT

indicative of potential adulteration and can be useful in detecting specimens that have been tampered with [161]. Unfortunately, urine drug testing is not effective in estimating serum drug concentrations or

SC

RI

assessing drug efficacy.

NU

Figure 4: EDDP Urine excretion window at steady state

MA

Pt XY 4500 4000

D

EDDP ng/mL

3500

2000 1500 1000 500 0

AC CE P

2500

TE

3000

60.00 50.00

UEW

40.00 30.00

problem samples

20.00

pH and UCr

5000

10.00 0.00

date of analysis EDDP

Creatinine

PH

Legend: *UEW urinary excretion window. The green lines represent the 2-ethylidene-1,5-dimethyl-3,3diphenylpyrrolidine (EDDP) excretion window.

In light of the limitations associated with urine drug testing, monitoring of pain management patients through serum or plasma drug measurements has been advocated. Therapeutic drug monitoring (TDM)involves the measurement of serum or plasma drug levels at timed intervals to determine whether steady state drug concentrations are within the therapeutic or expected range given the drug dosage [163].

29

ACCEPTED MANUSCRIPT TDM can be used by clinicians to explain drug toxicity or lack of efficacy, and can aid in adjusting drug doses or in choosing specific medications [163]. A major advantage of plasma drug measurement is that it integrates many factors affecting drug metabolism, as outlined in Figure 2. Half-life measurement can be a

PT

functional marker of the cumulative effect of pathophysiological and drug-drug interactions and provide an overall picture of the specific drug disposition and pharmacodynamics in the individual patient. However,

RI

TDM requires the collection of serial blood samples from patients, which can be difficult and inconvenient

SC

[160, 163]. Timing of sample collection also becomes an important consideration, depending on whether peak or trough measurements (or both) are to be monitored. The time window within which a particular drug or its metabolite can be detected in serum is an important consideration for sample collection. To

NU

assist in appropriate clinical interpretation, knowledge of tolerance and pharmacokinetics of particular

MA

drug(s) under investigation is essential.

D

Summary and Conclusions

TE

The experience of chronic pain is one of the commonest reasons individuals seek medical attention, making the management of chronic pain a major issue in clinical practice. Left untreated, chronic pain can lead to

AC CE P

diminished quality of life and socioeconomic difficulties. Opioids continue to be the mainstay of chronic pain management. However, opioid use can lead to abuse, diversion and physical dependence, meaning that opioids may not be suitable for all patients. An individual’s risk for abuse should be considered, by way of a detailed history, before opioids are prescribed. Several non-opioid based therapies are now potentially available for these patients, such as triptans, ziconotides, as well as novel therapies, such as treatment with cannabinoids and, in the not-so-distant future, gene therapy or epigenetic-based approaches. Adjuvant therapies with antidepressants, benzodiazepines or anticonvulsants can also be useful in managing pain, particularly in the treatment of anxiety and depression, which are commonly associated with chronic pain. Genetic variability can have profound effects on drug metabolism and contribute to the inter-individual diversity in responses to pain medications. However, there is a paucity of evidence for the benefits of pharmacogenetic testing in the context of pain management. Drug metabolism and responses are affected by many factors, including pharmacogenetics, with genetics offering only a partial explanation of an individual’s response.

30

ACCEPTED MANUSCRIPT At this point in time, therapeutic drug monitoring, through plasma drug measurements, may be more useful than pharmacogenetics in preventing adverse drug reactions to pain medications, while ensuring effective analgesia. Definitive, mass spectrometry based methods, capable of measuring parent drug and

PT

metabolite levels, are the most useful assays for this purpose. Drug half-life can be a functional marker of the cumulative effect of drug-drug interactions as well as pathophysiological interactions that may affect

RI

the treatment drug’s disposition. Currently, monitoring of pain management patients, if performed at all, is

SC

largely through urine drug measurements, which do not correlate with serum drug concentrations therefore limiting their use in monitoring efficacy and toxicity. Direct determination of serum or plasma drug levels may provide a more clinically useful, if logistically more difficult, avenue for optimization of

AC CE P

TE

D

MA

NU

drug therapy, particularly in the context of chronic pain management.

31

ACCEPTED MANUSCRIPT Reference List

PT

1. Bonica JJ. The need of a taxonomy. Pain. 1979;6:247-8.

RI

2. Furlan AD, Reardon R, Weppler C, Group NOUG. Opioids for chronic noncancer pain: A new canadian practice guideline. CMAJ Canadian Medical Association Journal. 2010;182:923-30.

SC

3. Hardt J, Jacobsen C, Goldberg J, Nickel R, Buchwald D. Prevalence of chronic pain in a representative sample in the united states. Pain Medicine. 2008;9:803-12.

NU

4. Organization WH. Cancer pain relief with a guide to opioid availability. 1996.

MA

5. Kroenke K, Krebs E, Wu J, Bair MJ, Damush T, Chumbler N, et al. Stepped care to optimize pain care effectiveness (scope) trial study design and sample characteristics. Contemporary Clinical Trials. 2013;34:270-81.

D

6. Treede RD, Jensen TS, Campbell JN, Cruccu G, Dostrovsky JO, Griffin JW, et al. Neuropathic pain: Redefinition and a grading system for clinical and research purposes. Neurology. 2008;70:1630-5.

TE

7. Gilron I, Watson CP, Cahill CM, Moulin DE. Neuropathic pain: A practical guide for the clinician. CMAJ Canadian Medical Association Journal. 2006;175:265-75.

AC CE P

8. Kroenke K, Krebs EE, Bair MJ. Pharmacotherapy of chronic pain: A synthesis of recommendations from systematic reviews. General Hospital Psychiatry. 2009;31:206-19. 9. Dubin AE, Patapoutian A. Nociceptors: The sensors of the pain pathway. Journal of Clinical Investigation. 2010;120:3760-72. 10. Tyrer S. Psychosomatic pain. British Journal of Psychiatry. 2006;188:91-3. 11. Arendt-Nielsen L, Svensson P. Referred muscle pain: Basic and clinical findings. Clinical Journal of Pain. 2001;17:11-9. 12. Kooijman CM, Dijkstra PU, Geertzen JH, Elzinga A, van der Schans CP. Phantom pain and phantom sensations in upper limb amputees: An epidemiological study. Pain. 2000;87:33-41. 13. Carr DB, Goudas LC. Acute pain. Lancet. 1999;353:2051-8. 14. Schopflocher D, Taenzer P, Jovey R. The prevalence of chronic pain in canada. Pain Research & Management. 2011;16:445-50. 15. Pergolizzi J, Ahlbeck K, Aldington D, Alon E, Coluzzi F, Dahan A, et al. The development of chronic pain: Physiological change necessitates a multidisciplinary approach to treatment. . Current Medical Research & Opinion. 2013;29:1127-35.

32

ACCEPTED MANUSCRIPT 16. Ricci JA, Stewart WF, Chee E, Leotta C, Foley K, Hochberg MC. Back pain exacerbations and lost productive time costs in united states workers. Spine. 2006;31:3052-60.

PT

17. Eriksen J, Ekholm O, Sjogren P, Rasmussen NK. Development of and recovery from long-term pain. A 6year follow-up study of a cross-section of the adult danish population. Pain. 2004;108:154-62.

RI

18. Elliott AM, Smith BH, Penny KI, Smith WC, Chambers WA. The epidemiology of chronic pain in the community. Lancet. 1999;354:1248-52.

SC

19. Kuehn BM. Opioid prescriptions soar: Increase in legitimate use as well as abuse. JAMA. 2007;297:24951.

NU

20. Prevention CfDCa. Selected prescription drug classes used in the past month, by sex and age years 1988-1994 through 2007-2010. http://wwwcdcgov/nchs/data/hus/hus12pdf#092. 2012 ed: Centers for Disease Control and Prevention; 2012. p. table 92.

MA

21. Stamer UM, Stuber F. Genetic factors in pain and its treatment. Current Opinion in Anaesthesiology. 2007;20:478-84.

D

22. Stamer UM, Zhang L, Stuber F. Personalized therapy in pain management: Where do we stand? Pharmacogenomics. 2010;11:843-64.

AC CE P

TE

23. Klepstad P, Rakvag TT, Kaasa S, Holthe M, Dale O, Borchgrevink PC, et al. The 118 a > g polymorphism in the human mu-opioid receptor gene may increase morphine requirements in patients with pain caused by malignant disease. Acta Anaesthesiologica Scandinavica. 2004;48:1232-9. 24. Lalovic B, Kharasch E, Hoffer C, Risler L, Liu-Chen LY, Shen DD. Pharmacokinetics and pharmacodynamics of oral oxycodone in healthy human subjects: Role of circulating active metabolites. Clinical Pharmacology & Therapeutics. 2006;79:461-79. 25. Kirchheiner J, Schmidt H, Tzvetkov M, Keulen JT, Lotsch J, Roots I, et al. Pharmacokinetics of codeine and its metabolite morphine in ultra-rapid metabolizers due to cyp2d6 duplication. Pharmacogenomics Journal. 2007;7:257-65. 26. Svetlik S, Hronova K, Bakhouche H, Matouskova O, Slanar O. Pharmacogenetics of chronic pain and its treatment. . Mediators of Inflammation. 2013;2013:864319. 27. Rendic S, Di Carlo FJ. Human cytochrome p450 enzymes: A status report summarizing their reactions, substrates, inducers, and inhibitors. Drug Metabolism Reviews. 1997;29:413-580. 28. Eichelbaum M, Ingelman-Sundberg M, Evans WE. Pharmacogenomics and individualized drug therapy. Annual Review of Medicine. 2006;57:119-37. 29. Wang B, Yang LP, Zhang XZ, Huang SQ, Bartlam M, Zhou SF. New insights into the structural characteristics and functional relevance of the human cytochrome p450 2d6 enzyme. Drug Metabolism Reviews. 2009;41:573-643.

33

ACCEPTED MANUSCRIPT 30. Zhou SF, Liu JP, Chowbay B. Polymorphism of human cytochrome p450 enzymes and its clinical impact. Drug Metabolism Reviews. 2009;41:89-295.

RI

PT

31. Johansson I, Lundqvist E, Bertilsson L, Dahl ML, Sjoqvist F, Ingelman-Sundberg M. Inherited amplification of an active gene in the cytochrome p450 cyp2d locus as a cause of ultrarapid metabolism of debrisoquine. Proceedings of the National Academy of Sciences of the United States of America. 1993;90:11825-9.

SC

32. Yeo RK, Rostami-Hodjegan A, Tucker GT. Abundance of cytochromes p450 in human liver: A metaanalysis. Proceedings of The British Pharmacological Society. 2004; http://www.pa2online.org/Vol1Issue4abst225P.html.

NU

33. Gasche Y, Daali Y, Fathi M, Chiappe A, Cottini S, Dayer P, et al. Codeine intoxication associated with ultrarapid cyp2d6 metabolism. New England Journal of Medicine. 2004;351:2827-31.

MA

34. Koren G, Cairns J, Chitayat D, Gaedigk A, Leeder SJ. Pharmacogenetics of morphine poisoning in a breastfed neonate of a codeine-prescribed mother. Lancet. 2006;368:704.

D

35. Canada H. Important safety information on tylenol with codeine in nursing mothers and ultra-rapid metabolizers of codeine - for health professionals. 2008.

AC CE P

TE

36. FDA warning on codeine use by nursing mothers. 2007. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/2007/ucm108968.htm. Accessed 21 January 2014 37. Pilotto A, Franceschi M, Vitale DF, Zaninelli A, Masotti G, Rengo F, et al. Upper gastrointestinal symptoms and therapies in elderly out-patients, users of non-selective nsaids or coxibs. Alimentary Pharmacology & Therapeutics. 2005;22:147-55. 38. Visser LE, van Schaik RH, van VM, Trienekens PH, De Smet PA, Vulto AG, et al. Allelic variants of cytochrome p450 2c9 modify the interaction between nonsteroidal anti-inflammatory drugs and coumarin anticoagulants. Clinical Pharmacology & Therapeutics. 2005;77:479-85. 39. Wadelius M, Chen LY, Lindh JD, Eriksson N, Ghori MJ, Bumpstead S, et al. The largest prospective warfarin-treated cohort supports genetic forecasting.[erratum appears in blood. 2014 feb 13;123(7):1113]. Blood. 2009;113:784-92. 40. Kirchheiner J, Stormer E, Meisel C, Steinbach N, Roots I, Brockmoller J. Influence of cyp2c9 genetic polymorphisms on pharmacokinetics of celecoxib and its metabolites. Pharmacogenetics. 2003;13:473-80. 41. Pilotto A, Seripa D, Franceschi M, Scarcelli C, Colaizzo D, Grandone E, et al. Genetic susceptibility to nonsteroidal anti-inflammatory drug-related gastroduodenal bleeding: Role of cytochrome p450 2c9 polymorphisms. Gastroenterology. 2007;133:465-71.

34

ACCEPTED MANUSCRIPT 42. Zwisler ST, Enggaard TP, Noehr-Jensen L, Mikkelsen S, Verstuyft C, Becquemont L, et al. The antinociceptive effect and adverse drug reactions of oxycodone in human experimental pain in relation to genetic variations in the oprm1 and abcb1 genes. Fundamental & Clinical Pharmacology. 2010;24:517-24.

PT

43. Campa D, Gioia A, Tomei A, Poli P, Barale R. Association of abcb1/mdr1 and oprm1 gene polymorphisms with morphine pain relief. Clinical Pharmacology & Therapeutics. 2008;83:559-66.

SC

RI

44. Levran O, O'Hara K, Peles E, Li D, Barral S, Ray B, et al. Abcb1 (mdr1) genetic variants are associated with methadone doses required for effective treatment of heroin dependence. Human Molecular Genetics. 2008;17:2219-27.

NU

45. Park HJ, Shinn HK, Ryu SH, Lee HS, Park CS, Kang JH. Genetic polymorphisms in the abcb1 gene and the effects of fentanyl in koreans. Clinical Pharmacology & Therapeutics. 2007;81:539-46.

MA

46. Lee YS, Kim H, Wu TX, Wang XM, Dionne RA. Genetically mediated interindividual variation in analgesic responses to cyclooxygenase inhibitory drugs. Clinical Pharmacology & Therapeutics. 2006;79:407-18.

D

47. Janicki PK, Schuler G, Francis D, Bohr A, Gordin V, Jarzembowski T, et al. A genetic association study of the functional a118g polymorphism of the human mu-opioid receptor gene in patients with acute and chronic pain. Anesthesia & Analgesia. 2006;103:1011-7.

AC CE P

TE

48. Fillingim RB, Kaplan L, Staud R, Ness TJ, Glover TL, Campbell CM, et al. The a118g single nucleotide polymorphism of the mu-opioid receptor gene (oprm1) is associated with pressure pain sensitivity in humans. Journal of Pain. 2005;6:159-67. 49. Landau R, Kern C, Columb MO, Smiley RM, Blouin JL. Genetic variability of the mu-opioid receptor influences intrathecal fentanyl analgesia requirements in laboring women. Pain. 2008;139:5-14. 50. Rakvag TT, Klepstad P, Baar C, Kvam TM, Dale O, Kaasa S, et al. The val158met polymorphism of the human catechol-o-methyltransferase (comt) gene may influence morphine requirements in cancer pain patients. Pain. 2005;116:73-8. 51. Reyes-Gibby CC, Shete S, Rakvag T, Bhat SV, Skorpen F, Bruera E, et al. Exploring joint effects of genes and the clinical efficacy of morphine for cancer pain: Oprm1 and comt gene. Pain. 2007;130:25-30. 52. Chen JF, Aloyo VJ, Weiss B. Continuous treatment with the d2 dopamine receptor agonist quinpirole decreases d2 dopamine receptors, d2 dopamine receptor messenger rna and proenkephalin messenger rna, and increases mu opioid receptors in mouse striatum. Neuroscience. 1993;54:669-80. 53. Steiner H, Gerfen CR. Role of dynorphin and enkephalin in the regulation of striatal output pathways and behavior. Experimental Brain Research. 1998;123:60-76. 54. Ross JR, Riley J, Taegetmeyer AB, Sato H, Gretton S, du Bois RM, et al. Genetic variation and response to morphine in cancer patients: Catechol-o-methyltransferase and multidrug resistance-1 gene polymorphisms are associated with central side effects. Cancer. 2008;112:1390-403.

35

ACCEPTED MANUSCRIPT 55. Kadiev E, Patel V, Rad P, Thankachan L, Tram A, Weinlein M, et al. Role of pharmacogenetics in variable response to drugs: Focus on opioids. Expert Opinion On Drug Metabolism & Toxicology. 2008;4:77-91.

PT

56. Fishbain DA, Fishbain D, Lewis J, Cutler RB, Cole B, Rosomoff HL, et al. Genetic testing for enzymes of drug metabolism: Does it have clinical utility for pain medicine at the present time? A structured review. Pain Medicine. 2004;5:81-93.

RI

57. Flockhart DA. Drug interactions: Cytochrome p450 drug interaction. 2014. http://medicine.iupui.edu/clinpharm/ddis/clinical-table/ Accessed 28 January 2014

SC

58. Ablin JN, Buskila D. Personalized treatment of pain. [review]. Current Rheumatology Reports. 2013;15:298.

NU

59. Cherny N, Ripamonti C, Pereira J, Davis C, Fallon M, McQuay H, et al. Strategies to manage the adverse effects of oral morphine: An evidence-based report. Journal of Clinical Oncology. 2001;19:2542-54.

MA

60. Rane A, Sawe J, Dahlstrom B, Paalzow L, Kager L. Pharmacological treatment of cancer pain with special reference to the oral use of morphine. Acta Anaesthesiologica Scandinavica. 1982;Supplementum.:74-103.

D

61. World Health Organization Cancer pain relief. 1986. http://www.whocancerpain.wisc.edu/?q=node/169 Accessed 21 January 2014

TE

62. Vargas-Schaffer G. Is the who analgesic ladder still valid? Twenty-four years of experience. Canadian Family Physician. 2010;56:514-7.

AC CE P

63. Varrassi G. Severe chronic pain--the reality of treatment in europe. Current Medical Research & Opinion. 2011;27:2063-4. 64. Rollins DE, von BC, Glaumann H, Moldeus P, Rane A. Acetaminophen: Potentially toxic metabolite formed by human fetal and adult liver microsomes and isolated fetal liver cells. Science. 1979;205:1414-6. 65. Mitchell JR, Jollow DJ, Potter WZ, Davis DC, Gillette JR, Brodie BB. Acetaminophen-induced hepatic necrosis. I. Role of drug metabolism. Journal of Pharmacology & Experimental Therapeutics. 1973;187:18594. 66. You K. Salicylate and mitochondrial injury in reye's syndrome. Science. 1983;221:163-5. 67. Reye RDK, Morgan G, Baral J. Encephalopathy and fatty degeneration of the viscera a disease entity on childhood. Lancet. 1963;2(7311):749-52. 68. Grosser T, Fries S, FitzGerald GA. Biological basis for the cardiovascular consequences of cox-2 inhibition: Therapeutic challenges and opportunities. Journal of Clinical Investigation. 2006;116:4-15. 69. Mukherjee D. Does a coxib-associated thrombotic risk limit the clinical use of the compounds as analgesic anti-inflammatory drugs? Arguments in favor. Thrombosis & Haemostasis. 2006;96:407-12.

36

ACCEPTED MANUSCRIPT 70. Antman EM, Bennett JS, Daugherty A, Furberg C, Roberts H, Taubert KA, et al. Use of nonsteroidal antiinflammatory drugs: An update for clinicians: A scientific statement from the american heart association. Circulation. 2007;115:1634-42.

RI

72. Portenoy RK. Treatment of cancer pain. Lancet. 2011;377:2236-47.

PT

71. Jones VM. Acetaminophen injection: A review of clinical information. [review]. Journal of Pain & Palliative Care Pharmacotherapy. 2011;25:340-9.

SC

73. Sawe J, Svensson JO, Rane A. Morphine metabolism in cancer patients on increasing oral doses--no evidence for autoinduction or dose-dependence. British Journal of Clinical Pharmacology. 1983;16:85-93.

NU

74. Koch T, Hollt V. Role of receptor internalization in opioid tolerance and dependence. Pharmacology & Therapeutics. 2008;117:199-206.

MA

75. Ferguson SS. Evolving concepts in g protein-coupled receptor endocytosis: The role in receptor desensitization and signaling. Pharmacological Reviews. 2001;53:1-24.

D

76. von Zastrow, Svingos A, Haberstock-Debic H, Evans C. Regulated endocytosis of opioid receptors: Cellular mechanisms and proposed roles in physiological adaptation to opiate drugs. Current Opinion in Neurobiology. 2003;13:348-53.

TE

77. Bailey CP, Connor M. Opioids: Cellular mechanisms of tolerance and physical dependence. Current Opinion in Pharmacology. 2005;5:60-8.

AC CE P

78. Mercadante S. Prospects and challenges in opioid analgesia for pain management. Current Medical Research & Opinion. 2011;27:1741-3. 79. Manchikanti L, Ailinani H, Koyyalagunta D, Datta S, Singh V, Eriator I, et al. A systematic review of randomized trials of long-term opioid management for chronic non-cancer pain. Pain Physician. 2011;14:91-121. 80. Al-Sayed AA, Al-Numay AM. Update and review on the basics of pain management. Neurosciences. 2011;16:203-12. 81. Martin WR. Pharmacology of opioids. Pharmacological Reviews. 1983;35:283-323. 82. Bohnert AS, Valenstein M, Bair MJ, Ganoczy D, McCarthy JF, Ilgen MA, et al. Association between opioid prescribing patterns and opioid overdose-related deaths. JAMA. 2011;305:1315-21. 83. Chang G, Chen L, Mao J. Opioid tolerance and hyperalgesia. Medical Clinics of North America. 2007;91:199-211. 84. Jamison RN, Serraillier J, Michna E. Assessment and treatment of abuse risk in opioid prescribing for chronic pain. Pain Res Treat. 2011;2011:941808. 85. Savage SR. Management of opioid medications in patients with chronic pain and risk of substance misuse. Current Psychiatry Reports. 2009;11:377-84.

37

ACCEPTED MANUSCRIPT 86. Bair MJ, Robinson RL, Katon W, Kroenke K. Depression and pain comorbidity: A literature review. Archives of Internal Medicine. 2003;163:2433-45. 87. Cupp M. New oxycodone formulation: Oxyneo. Pharmaacist's Letter / Prescriber's Letter2012.

PT

88. Houde RW. Analgesic effectiveness of the narcotic agonist-antagonists. British Journal of Clinical Pharmacology. 1979;7:297S-308S.

SC

RI

89. Vallejo R, Barkin RL, Wang VC. Pharmacology of opioids in the treatment of chronic pain syndromes. Pain Physician. 2011;14:E343-E60.

NU

90. Hutchinson MR, Menelaou A, Foster DJ, Coller JK, Somogyi AA. Cyp2d6 and cyp3a4 involvement in the primary oxidative metabolism of hydrocodone by human liver microsomes. British Journal of Clinical Pharmacology. 2004;57:287-97.

MA

91. Sansone RA, Sansone LA. Tramadol: Seizures, serotonin syndrome, and coadministered antidepressants. Psychiatry (Edgmont). 2009;6:17-21.

D

92. Stahl SM. Mechanism of action of serotonin selective reuptake inhibitors. Serotonin receptors and pathways mediate therapeutic effects and side effects. Journal of Affective Disorders. 1998;51:215-35.

TE

93. Nelson EM, Philbrick AM. Avoiding serotonin syndrome: The nature of the interaction between tramadol and selective serotonin reuptake inhibitors. Annals of Pharmacotherapy. 2012;46:1712-6.

AC CE P

94. Dayer P, Desmeules J, Collart L. [pharmacology of tramadol]. [french]. Drugs. 1997;53:Suppl-24. 95. Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clinical Pharmacokinetics. 2004;43:879-923. 96. Sawe J, Dahlstrom B, Paalzow L, Rane A. Morphine kinetics in cancer patients. Clinical Pharmacology & Therapeutics. 1981;30:629-35. 97. Cregg R, Russo G, Gubbay A, Branford R, Sato H. Pharmacogenetics of analgesic drugs. British Journal of Pain. 2013;7:189-206. 98. Berkenstadt H, Segal E, Mayan H, Almog S, Rotenberg M, Perel A, et al. The pharmacokinetics of morphine and lidocaine in critically ill patients. Intensive Care Medicine. 1999;25:110-2. 99. Smith MT. Neuroexcitatory effects of morphine and hydromorphone: Evidence implicating the 3glucuronide metabolites. Clinical & Experimental Pharmacology & Physiology. 2000;27:524-8. 100. Mather LE. Clinical pharmacokinetics of fentanyl and its newer derivatives. Clinical Pharmacokinetics. 1983;8:422-46. 101. Reynolds L, Rauck R, Webster L, DuPen S, Heinze E, Portenoy R, et al. Relative analgesic potency of fentanyl and sufentanil during intermediate-term infusions in patients after long-term opioid treatment for chronic pain. Pain. 2004;110:182-8.

38

ACCEPTED MANUSCRIPT 102. Kosek E, Jensen KB, Lonsdorf TB, Schalling M, Ingvar M. Genetic variation in the serotonin transporter gene (5-httlpr, rs25531) influences the analgesic response to the short acting opioid remifentanil in humans. Molecular Pain. 2009;5:37.

PT

103. Burkle H, Dunbar S, Van AH. Remifentanil: A novel, short-acting, mu-opioid. [review] [52 refs]. Anesthesia & Analgesia. 1996;83:646-51.

SC

RI

104. Anderson D. A review of systemic opioids commonly used for labor pain relief. [review][erratum appears in j midwifery womens health. 2011 jul-aug;56(4):411-8]. Journal of Midwifery & Women's Health. 2011;56:222-39.

NU

105. Leong WL, Sng BL, Sia AT. A comparison between remifentanil and meperidine for labor analgesia: A systematic review. [review]. Anesthesia & Analgesia. 2011;113:818-25.

MA

106. Kan RE, Hughes SC, Rosen MA, Kessin C, Preston PG, Lobo EP. Intravenous remifentanil: Placental transfer, maternal and neonatal effects. Anesthesiology. 1998;88:1467-74. 107. Fredheim OM, Borchgrevink PC, Klepstad P, Kaasa S, Dale O. Long term methadone for chronic pain: A pilot study of pharmacokinetic aspects. European Journal of Pain. 2007;11:599-604.

TE

D

108. Lugo RA, Satterfield KL, Kern SE. Pharmacokinetics of methadone. Journal of Pain & Palliative Care Pharmacotherapy. 2005;19:13-24.

AC CE P

109. Eap CB, Buclin T, Baumann P. Interindividual variability of the clinical pharmacokinetics of methadone: Implications for the treatment of opioid dependence. Clinical Pharmacokinetics. 2002;41:1153-93. 110. Kapur BM, Hutson JR, Chibber T, Luk A, Selby P. Methadone: A review of drug-drug and pathophysiological interactions. Critical Reviews in Clinical Laboratory Sciences. 2011;48:171-95. 111. Perrin-Terrin A, Pathak A, Lapeyre-Mestre M. Qt interval prolongation: Prevalence, risk factors and pharmacovigilance data among methadone-treated patients in france. Fundamental & Clinical Pharmacology. 2011;25:503-10. 112. Johnson RE, Fudala PJ, Payne R. Buprenorphine: Considerations for pain management. Journal of Pain & Symptom Management. 2005;29:297-326. 113. Pergolizzi J, Aloisi AM, Dahan A, Filitz J, Langford R, Likar R, et al. Current knowledge of buprenorphine and its unique pharmacological profile. Pain Practice. 2010;10:428-50. 114. Leow KP, Cramond T, Smith MT. Pharmacokinetics and pharmacodynamics of oxycodone when given intravenously and rectally to adult patients with cancer pain. Anesthesia & Analgesia. 1995;80:296-302. 115. Sloan P. Review of oral oxymorphone in the management of pain. Ther Clin Risk Manag. 2008;4:77787. 116. Batterman RC. Clinical effectiveness and safety of a new synthetic analgesic drug, demerol. Archives of Internal Medicine. 1943;71:345-56.

39

ACCEPTED MANUSCRIPT 117. Latta KS, Ginsberg B, Barkin RL. Meperidine: A critical review. American Journal of Therapeutics. 2002;9:53-68.

PT

118. Saravanakumar K, Garstang JS, Hasan K. Intravenous patient-controlled analgesia for labour: A survey of uk practice. International Journal of Obstetric Anesthesia. 2007;16:221-5.

RI

119. Tveit TO, Halvorsen A, Rosland JH. Analgesia for labour: A survey of norwegian practice - with a focus on parenteral opioids. Acta Anaesthesiologica Scandinavica. 2009;53:794-9.

SC

120. Prommer E. Levorphanol: The forgotten opioid. Supportive Care in Cancer. 2007;15:259-64.

NU

121. Sansone RA, Sansone LA. Pain, pain, go away: Antidepressants and pain management. Psychiatry (Edgmont). 2008;5:16-9.

MA

122. Gureje O, Von KM, Simon GE, Gater R. Persistent pain and well-being: A world health organization study in primary care. JAMA. 1998;280:147-51.

D

123. Eardley W, Toth C. An open-label, non-randomized comparison of venlafaxine and gabapentin as monotherapy or adjuvant therapy in the management of neuropathic pain in patients with peripheral neuropathy. J Pain Res. 2010;1:33-49.

TE

124. Moulin DE, Clark AJ, Gilron I, Ware MA, Watson CP, Sessle BJ, et al. Pharmacological management of chronic neuropathic pain - consensus statement and guidelines from the canadian pain society. Pain Research & Management. 2007;12:13-21.

AC CE P

125. Jensen TS. Anticonvulsants in neuropathic pain: Rationale and clinical evidence. European Journal of Pain. 2002;6:A61-8. 126. Bagheri SC, Farhidvash F, Perciaccante VJ. Diagnosis and treatment of patients with trigeminal neuralgia. J Am Dent Assoc. 2004;135:1713-37. 127. Dolder CR, Nealy KL. The efficacy and safety of newer anticonvulsants in patients with dementia. Drugs & Aging. 2012;29:627-37. 128. Rose MA, Kam PC. Gabapentin: Pharmacology and its use in pain management. Anaesthesia. 2002;57:451-62. 129. Ward KE, Archambault R, Mersfelder TL. Severe adverse skin reactions to nonsteroidal antiinflammatory drugs: A review of the literature. American Journal of Health-System Pharmacy. 2010;67:206-13. 130. Gentile G, Borro M, Simmaco M, Missori S, Lala N, Martelletti P. Gene polymorphisms involved in triptans pharmacokinetics and pharmacodynamics in migraine therapy. Expert Opinion On Drug Metabolism & Toxicology. 2011;7:39-47. 131. Loder E. Triptan therapy in migraine. New England Journal of Medicine. 2010;363:63-70.

40

ACCEPTED MANUSCRIPT 132. Gawel MJ, Worthington I, Maggisano A. A systematic review of the use of triptans in acute migraine. Canadian Journal of Neurological Sciences. 2001;28:30-41.

PT

133. Dellemijn PL, Fields HL. Do benzodiazepines have a role in chronic pain management?. Pain. 1994;57:137-52.

RI

134. Manzanares J, Julian M, Carrascosa A. Role of the cannabinoid system in pain control and therapeutic implications for the management of acute and chronic pain episodes. Curr Neuropharmacol. 2006;4:23957.

SC

135. Walker JM, Huang SM. Cannabinoid analgesia. Pharmacology & Therapeutics. 2002;95:127-35.

NU

136. Leung L. Cannabis and its derivatives: Review of medical use. Journal of the American Board of Family Medicine: JABFM. 2011;24:452-62.

MA

137. Abrams DI, Jay CA, Shade SB, Vizoso H, Reda H, Press S, et al. Cannabis in painful hiv-associated sensory neuropathy: A randomized placebo-controlled trial. Neurology. 2007;68:515-21.

D

138. Ellis RJ, Toperoff W, Vaida F, van den BG, Gonzales J, Gouaux B, et al. Smoked medicinal cannabis for neuropathic pain in hiv: A randomized, crossover clinical trial. Neuropsychopharmacology. 2009;34:672-80.

TE

139. Rog DJ, Nurmikko TJ, Friede T, Young CA. Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis. Neurology. 2005;65:812-9.

AC CE P

140. Vaney C, Heinzel-Gutenbrunner M, Jobin P, Tschopp F, Gattlen B, Hagen U, et al. Efficacy, safety and tolerability of an orally administered cannabis extract in the treatment of spasticity in patients with multiple sclerosis: A randomized, double-blind, placebo-controlled, crossover study. Multiple Sclerosis. 2004;10:417-24. 141. Zajicek JP, Sanders HP, Wright DE, Vickery PJ, Ingram WM, Reilly SM, et al. Cannabinoids in multiple sclerosis (cams) study: Safety and efficacy data for 12 months follow up. Journal of Neurology, Neurosurgery & Psychiatry. 2005;76:1664-9. 142. Ware M, Beaulieu P. Cannabinoids for the treatment of pain: An update on recent clinical trials. . Pain Research & Management. 2005;10:27A-30A. 143. de Vries M, van Rijckevorsel DC, Wilser-Smith OH, van Goor H. Dronabinol and chronic pain: Importance of mechanistic considerations. Expert Opinion on Pharmacotherapy. 2014;12:1-10. 144. Clark AJ, Lynch ME, Ware M, Beaulieu P, McGilveray IJ, Gourlay D. Guidelines for the use of cannabinoid compounds in chronic pain. [review]. Pain Research & Management. 2005;10:44A-6A. 145. Donoghue K, Doody GA, Murray RM, Jones PB, Morgan C, Dazzan P, et al. Cannabis use, gender and age of onset of schizophrenia: Data from the æsop study. Psychiatry Research. 2014;215:528-32. 146. Olivera BM. Conus peptides: Biodiversity-based discovery and exogenomics. Journal of Biological Chemistry. 2006;281:31173-7.

41

ACCEPTED MANUSCRIPT 147. Schmidtko A, Lotsch J, Freynhagen R, Geisslinger G. Ziconotide for treatment of severe chronic pain. Lancet. 2010;375:1569-77.

PT

148. Wermeling D, Drass M, Ellis D, Mayo M, McGuire D, O'Connell D, et al. Pharmacokinetics and pharmacodynamics of intrathecal ziconotide in chronic pain patients. Journal of Clinical Pharmacology. 2003;43:624-36.

RI

149. Geranton SM. Targeting epigenetic mechanisms for pain relief. Current Opinion in Pharmacology. 2012;12:35-41.

SC

150. Tajerian M, Alvarado S, Millecamps M, Dashwood T, Anderson KM, Haglund L, et al. DNA methylation of sparc and chronic low back pain. Molecular Pain. 2011;7:65.

NU

151. Uchida H, Sasaki K, Ma L, Ueda H. Neuron-restrictive silencer factor causes epigenetic silencing of kv4.3 gene after peripheral nerve injury. Neuroscience. 2010;166:1-4.

MA

152. Uchida H, Ma L, Ueda H. Epigenetic gene silencing underlies c-fiber dysfunctions in neuropathic pain. Journal of Neuroscience. 2010;30:4806-14.

D

153. Robak T. New nucleoside analogs for patients with hematological malignancies. Expert Opinion on Investigational Drugs. 2011;20:343-59.

TE

154. Bai G, Wei D, Zou S, Ren K, Dubner R. Inhibition of class ii histone deacetylases in the spinal cord attenuates inflammatory hyperalgesia. Molecular Pain. 2010;6:51.

AC CE P

155. Agrawal RP, Goswami J, Jain S, Kochar DK. Management of diabetic neuropathy by sodium valproate and glyceryl trinitrate spray: A prospective double-blind randomized placebo-controlled study. Diabetes Research & Clinical Practice. 2009;83:371-8. 156. Handy CR, Krudy C, Boulis N. Gene therapy: A potential approach for cancer pain. Pain Research and Treatment. 2011;2011;2011:987597. 157. Fink DJ, Wechuck J, Mata M, Glorioso JC, Goss J, Krisky D, et al. Gene therapy for pain: Results of a phase i clinical trial. Annals of Neurology. 2011;70:207-12. 158. Manchikanti L, Damron KS, McManus CD, Barnhill RC. Patterns of illicit drug use and opioid abuse in patients with chronic pain at initial evaluation: A prospective, observational study. Pain Physician. 2004;7:431-7. 159. Manchikanti L, Manchukonda R, Pampati V, Damron KS, Brandon DE, Cash KA, et al. Does random urine drug testing reduce illicit drug use in chronic pain patients receiving opioids? Pain Physician. 2006;9:123-9. 160. Manchikanti L, Atluri S, Trescot AM, Giordano J. Monitoring opioid adherence in chronic pain patients: Tools, techniques, and utility. Pain Physician. 2008;11:Suppl-80.

42

ACCEPTED MANUSCRIPT 161. Nafziger AN, Bertino JS, Jr. Utility and application of urine drug testing in chronic pain management with opioids. Clinical Journal of Pain. 2009;25:73-9.

PT

162. Kapur BM, Verjee Z, Anderson M, Geisbrecht E. Urinary eddp (methadone metabolite) as a marker of compliance and diversion of methadone. Therapeutic Drug Monitoring. 1995;17:388.

RI

163. Jannetto PJ, Bratanow NC. Pain management in the 21st century: Utilization of pharmacogenomics and therapeutic drug monitoring. Expert Opinion On Drug Metabolism & Toxicology. 2011;7:745-52.

SC

164. Levy G, Tsuchiya T. Salicylate accumulation kinetics in man. New England Journal of Medicine. 1972;287:430-2.

NU

165. Adams SS, McCullough KF, Nicholson JS. The pharmacological properties of ibuprofen, an antiinflammatory, analgesic and antipyretic agent. Archives Internationales de Pharmacodynamie et de Therapie. 1969;178:115-29.

MA

166. Alvan G, Orme M, Bertilsson L, Ekstrand R, Palmer L. Pharmacokinetics of indomethacin. Clinical Pharmacology & Therapeutics. 1975;18:364-73.

D

167. Morris ME, Levy G. Renal clearance and serum protein binding of acetaminophen and its major conjugates in humans. Journal of Pharmaceutical Sciences. 1984;73:1038-41.

TE

168. Davies NM, McLachlan AJ, Day RO, Williams KM. Clinical pharmacokinetics and pharmacodynamics of celecoxib: A selective cyclo-oxygenase-2 inhibitor. Clinical Pharmacokinetics. 2000;38:225-42.

AC CE P

169. Baselt R. Disposition of toxic drugs and chemicals in man. Ninth Edition ed. Seal Beach, California: Biomedical Publications; 2011. 170. Blaine JD. Buprenorphine: An alternative treatment for opioid dependence. In: Jack D.Blaine P, Ed. 5600 Fishers Lane, Rockville, MD 20857, USA: National Institute on Drug Abuse.; 1992. 171. Bullingham RE, McQuay HJ, Moore A, Bennett MR. Buprenorphine kinetics. Clinical Pharmacology & Therapeutics. 1980;28:667-72. 172. Han T, Harmatz JS, Greenblatt DJ, Martyn JA. Fentanyl clearance and volume of distribution are increased in patients with major burns. Journal of Clinical Pharmacology. 2007;47:674-80. 173. Parab PV, Ritschel WA, Coyle DE, Gregg RV, Denson DD. Pharmacokinetics of hydromorphone after intravenous, peroral and rectal administration to human subjects. Biopharmaceutics & Drug Disposition. 1988;9:187-99. 174. Ramirez J, Innocenti F, Schuetz EG, Flockhart DA, Relling MV, Santucci R, et al. Cyp2b6, cyp3a4, and cyp2c19 are responsible for the in vitro n-demethylation of meperidine in human liver microsomes. Drug Metabolism & Disposition. 2004;32:930-6. 175. Pond SM, Tong T, Benowitz NL, Jacob P, Rigod J. Presystemic metabolism of meperidine to normeperidine in normal and cirrhotic subjects. Clinical Pharmacology & Therapeutics. 1981;30:183-8.

43

ACCEPTED MANUSCRIPT 176. Schulz P, Turner-Tamiyasu K, Smith G, Giacomini KM, Blaschke TF. Amitriptyline disposition in young and elderly normal men. Clinical Pharmacology & Therapeutics. 1983;33:360-6.

PT

177. Stewart JJ, Berkel HJ, Parish RC, Simar MR, Syed A, Bocchini JA, Jr., et al. Single-dose pharmacokinetics of bupropion in adolescents: Effects of smoking status and gender. Journal of Clinical Pharmacology. 2001;41:770-8.

SC

RI

178. Rampono J, Kristensen JH, Hackett LP, Paech M, Kohan R, Ilett KF. Citalopram and demethylcitalopram in human milk; distribution, excretion and effects in breast fed infants. British Journal of Clinical Pharmacology. 2000;50:263-8.

NU

179. Rudorfer MV, Lane EA, Chang WH, Zhang MD, Potter WZ. Desipramine pharmacokinetics in chinese and caucasian volunteers. British Journal of Clinical Pharmacology. 1984;17:433-40. 180. Deepak P, Kumar TN. Duloxetine - pharmacological aspects. Int J Biol Med Res. 2011;2:589-92.

MA

181. Altamura AC, Moro AR, Percudani M. Clinical pharmacokinetics of fluoxetine. Clinical Pharmacokinetics. 1994;26:201-14.

D

182. Ellingrod VL, Perry PJ. Venlafaxine: A heterocyclic antidepressant. American Journal of Hospital Pharmacy. 1994;51:3033-46.

TE

183. Mesdjian E, Seree E, Charvet B, Mirrione A, Bourgarel-Rey V, Desobry A, et al. Metabolism of carbamazepine by cyp3a6: A model for in vitro drug interactions studies. Life Sciences. 1999;64:827-35.

AC CE P

184. Rawlins MD, Collste P, Bertilsson L, Palmer L. Distribution and elimination kinetics of carbamazepine in man. European Journal of Clinical Pharmacology. 1975;8:91-6. 185. McLean MJ. Clinical pharmacokinetics of gabapentin. Neurology. 1994;44:Suppl-22. 186. Chan V, Morris RG, Ilett KF, Tett SE. Population pharmacokinetics of lamotrigine. Therapeutic Drug Monitoring. 2001;23:630-5. 187. Bergen DC. Pharmacokinetics of phenytoin: Reminders and discoveries. Epilepsy Curr. 2009;9:102-4. 188. Cuttle L, Munns AJ, Hogg NA, Scott JR, Hooper WD, Dickinson RG, et al. Phenytoin metabolism by human cytochrome p450: Involvement of p450 3a and 2c forms in secondary metabolism and drug-protein adduct formation. Drug Metabolism & Disposition. 2000;28:945-50.

44

ACCEPTED MANUSCRIPT Legends:

PT

Figure 1: Cytochrome p450: Abundance and proportions b) CYP Abundance in human liver

Adapted from: Rendic S and Di Carlo FJ[27]

Adapted from Yeo et. al. [32]

SC

RI

a) Proportions of Drugs Metabolized by CYP 450 Enzymes

NU

Figure 2 Genetic and non-genetic factors effecting drug metabolism Adapted from Stamer et. al.[22]

MA

Figure 3 The WHO three-step analgesic ladder Adapted from [60, 61]

TE

D

Legend: The pain ladder shows a progressive rationale for stronger opioids. Before a new medication is introduced, it is important to wait until the previously administered drug has reached a “steady state” or full effect is achieved.

AC CE P

*Adjuvants: antidepressants, anticonvulsants, steroids

Figure 4: EDDP Urine excretion window at steady state Legend:

*UEW urinary excretion window. The green lines represent the 2-ethylidene-1,5-dimethyl-3,3diphenylpyrrolidine (EDDP) excretion window.

45

ACCEPTED MANUSCRIPT Table 1: Properties of NSAIDs and acetaminophen used in the management of chronic pain*. Protein

Administration

Binding

Oral

99%

Bioavailability#

Route of

Vd



Elimination

L/Kg

(h)

80-100%

Renal

0.1--0.2

2-3 (low doses)

99%

49-73%

Renal

0.14

0.9-2.5

98%

~100 %(oral), 80 to 90% (rectal)

Renal Fecal

0.4--1.2

5-10

PT

Common Route of

RI

Drug#

PT ED

MA

NU

SC

acetylsalicylic acid (aspirin ) [164]

Ibuprofen [165]

AC

CE

Oral

Indomethacin [166]

Oral

46

ACCEPTED MANUSCRIPT Acetaminophen [167] 20%

Oral

97%

~100%

Renal

0.25

1-4

Renal

5.7--7.0

11-16

SC

RI

PT

Oral

unknown

CE

PT ED

MA

NU

Celecoxib [168]

AC

# All chemical formulas and bioavailability are from their respective pages in Wikipedia; **[169]

47

ACCEPTED MANUSCRIPT Table 2: Properties of Opioid medications commonly used in the management of chronic pain. Protein

Bio -

Route of

Vd**

t½**

Administration*

Binding

availability #

elimination

L/Kg

(h)

CYP

RI

PT

Common Route of Drug#

NU

TD, B, IV

31%

SC

Buprenorphine [170, 171]

96%

PT ED

Codeine [89]

2B6>2D6

(oral)

CE

87%

AC

Morphine [89, 98]

20–40% (oral),

IV, IM, oral

35%

36–71% (rectally), 100%

Glucuronidation Renal (90%) 2-5

1.3-6.7 6.7

(major)

Biliary (10%) 2D6 (minor)

(IV/IM)

50

ACCEPTED MANUSCRIPT

Meperidine (Pethidine) [174,

Oral, IV, IM

45-64%

SC

80-90% (Oral, in cases

2B6, 3A4 &

hepatic

3.7-4.2

2-5 5

2C19(minor)

Renal

1.8-3.7

3-6 6

2D6

renal

2.4

4-12 12

glucuronidation

NU

of hepatic

RI

50–60% (Oral),

PT

175]

MA

impairment)

PT ED

Oxycodone [114]

45%

AC

Oral

CE

60-87%

Oxymorphone [115]

10% (oral), 40%

Oral

10-12%

(Intranasal), 100%

fecal

(IV, IM)

51

ACCEPTED MANUSCRIPT

PT

Remifentanyl

Not applicable

plasma

Given iv

i.v

1-20 20

proteins)

NU

Tramadol [89, 95]

Nonspecific esterases

minute

SC

(bound to

RI

70%

70-75%

Oral

15-20%

MA

(oral), 77%

renal

2.6-2.9

4.3-6.7 6.7

2D6>2B6, 3A4

PT ED

(rectal),

CE

100% (IM)

AC

# All chemical formulas and bioavailability are from their respective pages in Wikipedia; * depends on formulation; 2-3h 2 for immediate release, 5h for continuous release; TD transdermal; B buccal; IV intra venous; ** **[169]

52

ACCEPTED MANUSCRIPT Table 3: Antidepressants commonly used in pain management Bio availability #

Route of elimination

Vd* L/Kg

t½* (h)

CYP

Renal

6-10

8-51

2D6

Renal Fecal

40

4-24

2B6

PT

Protein Binding

Amitriptyline [176]

> 90%

Oral

85%

30–60% due to first pass metabolism

PT ED

MA

Oral

SC

RI

Common Route of Administration

NU

Drug#

??????

AC

CE

Bupropion [177]

53

ACCEPTED MANUSCRIPT

Oral

70-90%

Renal

12-16

25-40

2C19, 3A4, 2D6

73-92%

Renal

22-59

12-54

2D6

32% to 80%

Renal Fecal

17-26

8-17

1A2,2D6

80%

RI

50%

NU

SC

Oral

PT

Citalopram [178]

Duloxetine [180]

Oral

AC

CE

PT ED

MA

Desipramine [179]

>90%

54

ACCEPTED MANUSCRIPT Fluoxetine[181]

Oral

27%

Renal Fecal

PT

94%

72%

20-42

1-3 days

2D6

4-12

3-7

2D6, 3A4

RI

Oral

NU

SC

Venlafaxine [182]

renal

PT ED

MA

42±15%

AC

CE

# All chemical formulas and bioavailability data are from their respective pages in Wikipedia; *[169]

55

ACCEPTED MANUSCRIPT Table 4: Anticonvulsant drugs used in pain management Common Route of Administration

Protein Binding

Bio availability #

Oral

75%

??????

Route of elimination

Vd* L/Kg

t½* (h)

CYP

Renal

0.8-1.8

18 18-65

3A4

27-60% (inversely proportional to dose)

Renal

0.8-1.3

5 5-9

Not metabolized

55%

98%

Renal

0.9-1.3

12 12-62

glucuronidation

87%-93%

70-100% oral, 24.4% (rectal, IV)

Biliary

0.5-0.8

8--60

2C19

PT

Drug#

< 3%

Oral

PT ED

Oral

CE

Gabapentin [185]

MA

NU

SC

RI

Carbamazepine [183, 184]

AC

Lamotrigine [186]

Phenytoin [187, 188]

Oral, IV

# All chemical formulas and bioavailability data are from their respective pages in Wikipedia; **[169] 56

ACCEPTED MANUSCRIPT

Vd** L/Kg

t½** (h)

Renal Fecal

2.5-4

3-4 4

3A4, MAO*, 2D6

50%

Renal

2.1-3.3

3-7 7

3A4

20–30%

Renal

3-4.2

20-30 30

1A2, 2D6

Renal

2-3

5-6 6

Unknown CYP

Common Route of Administration

Protein Binding

Bio availability #

Oral

35%

70%

Oral

85%

Route of elimination

CYP

RI

Drug#

PT

Table 5: Triptans used in migraine therapy[130, 132]

MA

NU

SC

Almotriptan[131]

Oral

AC

CE

Frovatriptan[131]

PT ED

Eletriptan[131]

15%

Naratriptan[131]

Oral

20-30%

74%

57

ACCEPTED MANUSCRIPT Rizatriptan[131]

Oral

14-21%

Oral, nasal

25%

Renal

1.3-2.2

1.6-3.2 3.2

MAO, 1A2

Renal

1.3-4.6

1-4 4

MOA

Renal Fecal

7.0

1.6-3.8 3.8

1A2, MAO, 3A4

PT

14%

45%

RI

Oral

NU

SC

Sumatriptan[131]

PT ED

MA

15% (oral) 96% (s.c)

40% (oral)

AC

CE

Zolmitriptan[131]

# All chemical formulas and bioavailability data are from their respective pages in Wikipedia; *MAO – monoamine oxidase; **[169] **

58

ACCEPTED MANUSCRIPT

AC

CE

PT ED

MA

NU

SC

RI

PT

Graphical abstract

59

ACCEPTED MANUSCRIPT Highlights

TE

D

MA

NU

SC

RI

PT

We review different types of pain Pharmacogenetic of pain The mechanisms and enzymes involved in pain medications Pharmacogenetic and pharmacological properties of drugs used in the management of pain

AC CE P

-

60

Pharmacogenetics of chronic pain management.

The experience of chronic pain is one of the commonest reasons individuals seek medical attention, making the management of chronic pain a major issue...
1MB Sizes 2 Downloads 4 Views