Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/262939924

Pharmacogenetics of Chronic Pain Management.

Article  in  Clinical Biochemistry · June 2014


DOI: 10.1016/j.clinbiochem.2014.05.065 · Source: PubMed

CITATIONS READS

40 1,269

3 authors, including:

Prateek Lala Bhushan M Kapur


University of Toronto University of Toronto
8 PUBLICATIONS   403 CITATIONS    150 PUBLICATIONS   2,385 CITATIONS   

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Prateek Lala on 06 July 2014.

The user has requested enhancement of the downloaded file.


CLB-08771; No. of pages: 19; 4C:
Clinical Biochemistry xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Clinical Biochemistry
journal homepage: www.elsevier.com/locate/clinbiochem

Review

Pharmacogenetics of chronic pain management


Bhushan M. Kapur a,b,c,⁎, Prateek K. Lala b, Julie L.V. Shaw c
a
Department of Clinical Pathology, Sunnybrook Health Sciences Center, Toronto, Canada
b
Division of Clinical Pharmacology and Toxicology, The Hospital for Sick Children University of Toronto, Canada
c
Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Canada

a r t i c l e i n f o a b s t r a c t

Article history: Objective: The experience of chronic pain is one of the commonest reasons individuals seek medical atten-
Received 8 March 2014 tion, making the management of chronic pain a major issue in clinical practice. Drug metabolism and responses
Received in revised form 25 May 2014 are affected by many factors, with genetic variations offering only a partial explanation of an individual’s response.
Accepted 27 May 2014 There is a paucity of evidence for the benefits of pharmacogenetic testing in the context of pain management.
Available online xxxx
Design and methods: We reviewed the literature between 2000 and 2013, and references cited therein, using
various keywords related to pain management, pharmacology and pharmacogenetics.
Keywords:
Chronic pain
Results: Opioids continue to be the mainstay of chronic pain management. Several non-opioid based therapies,
Pain management such as treatment with cannabinoids, gene therapy and epigenetic-based approaches are now available for these
Pharmacokinetics patients. Adjuvant therapies with antidepressants, benzodiazepines or anticonvulsants can also be useful in man-
Pharmacogenetics aging pain. Currently, laboratory monitoring of pain management patients, if performed, is largely through urine
Opioids drug measurements.
NSAIDs Conclusions: Drug half-life calculations can be used as functional markers of the cumulative effect of pharma-
Triptans cogenetics and drug–drug interactions. Assessment of half-life and therapeutic effects may be more useful than ge-
netic 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.
© 2014 The Authors. The Canadian Society of Clinical Chemists. Published by Elsevier Inc. This is an open access
article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/3.0/).

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Pain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Chronic pain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Pharmacogenetics and pain management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Cytochrome P450 enzymes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
P-glycoprotein . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Cyclooxygenases 1 and 2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Opioid receptor μ . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Catechol-O-methyltransferase (COMT) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Clinical use of pharmacogenetics in pain management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Genetics and pain susceptibility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Chronic pain management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Drugs used in pain management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
NSAIDs and acetaminophen in pain management (Table 1) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Opioids and pain management (Table 2) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Issues with opioids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Specific opioids used in pain management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

⁎ Corresponding author at: Department of Clinical Pathology, Sunnybrook Health Science Centre, 2075 Bayview Ave, Toronto, Ontario, M4N 3M5, Canada. Fax: +1 905 849 4389.
E-mail address: b.kapur@utoronto.ca (B.M. Kapur).

http://dx.doi.org/10.1016/j.clinbiochem.2014.05.065
0009-9120/© 2014 The Authors. The Canadian Society of Clinical Chemists. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/3.0/).

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
2 B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx

Weak opioids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Codeine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Hydrocodone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Tramadol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Strong opioids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Morphine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Hydromorphone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Fentanyl . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Remifentanil . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Methadone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Buprenorphine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Oxycodone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Oxymorphone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Meperidine (pethidine) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Levorphanol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Other medications in the treatment of chronic pain . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Antidepressants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Anticonvulsants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Triptans . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Benzodiazepines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Cannabinoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Ziconotide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Novel therapies in pain management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Targeting epigenetic modifications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Gene therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Therapeutic drug monitoring in pain management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
Summary and conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 0

Introduction choice. In addition, novel therapies, including targeting of epigenetic


changes and gene therapy-based approaches are further broadening
According to the International Association for the Study of Pain future options for the treatment of chronic pain.
(IASP), pain is defined as an “unpleasant sensory and emotional experi-
ence associated with actual or potential tissue damage, or described in Methods
terms of such damage”. Several different subtypes of pain can be de-
scribed, based on their neuropsychological basis and duration, including We conducted an online systematic search for papers and related
neuropathic, nociceptive, psychogenic, referred, phantom, acute and abstracts published between 2000 and 2013 using the National Library
chronic [1]. Chronic pain is very common, with one in three Americans of Medicine database, PubMed. Using the following headings: “pain
and one in five Canadians reported to suffer from this problem [2,3]. It management medications”, “pain management drugs”, “chronic pain
is one of the most frequent reasons for individuals to seek medical management”, “pharmacogenetics and pain management”, and “drug
care. If untreated, chronic pain can lead to physical and social dysfunction monitoring and pain management”, we identified 6670 papers. The ab-
and diminished quality of life. stracts of all papers were reviewed and those that appeared to be rele-
Successful pain management provides adequate analgesia without vant to the subject were selected for further study. References cited
excessive adverse effects. Current pain management strategies largely therein, including original publications, were also selected for further
employ the use of the World Health Organization (WHO) pain ladder, review.
beginning with non-opioid medications, such as NSAIDs, progressing
to weak opioids, and culminating with strong opioids, particularly in Pain
cancer pain [4]. The WHO also recommends adjuvant therapy with an-
tidepressant medications to aid in reducing anxiety often associated Pain is the most common presenting physical symptom in primary
with chronic pain [4]. care, accounting for an enormous burden of patient suffering, quality
Management of pain can be complicated by lack of adherence, the of life, work and social disability, and health care and societal costs [5].
potential for abuse or dependence on the medications used and adverse Pain is defined as an unpleasant sensory and emotional experience
drug side effects. Many factors can influence drug disposition, including associated with actual or potential tissue damage [1]. However, the ex-
genetic variation, which can further complicate management of these perience of pain is highly subjective and idiosyncratic, and individuals
patients. develop pain thresholds through experiences of injury or pathology in
Genetic studies have identified several loci in which polymorphic early life [1].
changes can influence the pharmacodynamics and kinetics of analgesic Neuropathic pain results from actual damage to nerve fibers them-
drugs. Current patient monitoring in pain management (if performed at selves in the central or peripheral nervous system [6]. This damage can
all) is largely based on urine drug measurements. However, therapeutic lead to nerve dysfunction, causing numbness, weakness and/or loss of
drug monitoring (TDM) via plasma drug levels and half-life may prove reflexes. Common descriptions accompanying neuropathic pain include
more useful in monitoring patients prescribed pain medications to “burning”, “shooting” or “shock-like” sensations [7]. The most common
ensure efficacy while minimizing adverse drug reactions. TDM can causes of chronic neuropathic pain include diabetic neuropathy, post-
also be very useful for the identification of drug-related side effects herpetic neuralgia, fibromyalgia, lower back pain (radiculopathy due
and patient-reported lack of effect (e.g., tolerance). to disc herniation) and osteoarthritis [8].
Finally, non-opioid analgesics are often tried as options for manage- Nociceptive pain is caused by stimulation of specialized sensory
ment of pain, particularly in individuals where opioids are not a suitable neurons, called nociceptors, by noxious stimuli including extremes of

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx 3

temperature, or mechanical or chemical excitation. It is often catego- can be divided into two broad categories: those genes affecting pharma-
rized as somatic (derived from skin and deep tissue) or visceral cokinetics, and those affecting pharmacodynamics [22]. In the case of
(originating from internal organs). Due to high concentrations of pain management drugs, genes associated with altered pharmacokinet-
nociceptors in somatic tissues, chronic somatic pain is typically local- ics include those which encode members of the cytochrome P450 family
ized, and often results from degenerative or inflammatory processes. of enzymes, enzymes responsible for glucuronidation and drug trans-
Nociceptive pain is frequently described as stabbing, pricking, burning, porter proteins [21,22]. Genes encoding cyclooxygenase enzymes, the
throbbing or cramping [9]. opioid receptors and the enzyme catecholamine methyltransferase
Psychogenic pain refers to painful sensations caused or amplified by (COMT) can affect drug pharmacodynamics [21,22]. Světlík and col-
mental or emotional factors with no evident physical tissue damage leagues provide an up-to-date review of results from pharmacogenetic
[10]. Referred pain is perceived at a site adjacent to or distant from the studies of candidate genes in both the kinetic and dynamic domains [26].
site of painful stimulus; the mechanism for this process is not yet well
understood [11]. Phantom pain, a sub-type of referred pain, is the feeling
of painful sensations from a part of the body which has been lost, and Cytochrome P450 enzymes
from which the brain no longer receives signals. Individuals who have
undergone limb amputation often experience phantom pain [12]. The cytochrome P450 (CYP) enzymes comprise a category of
Acute and chronic are terms commonly used to describe the duration proteins whose biosynthesis is controlled by a large super-family of
of painful sensations, but are difficult to categorically define. In general, genes. Members of the CYP family play important roles in drug metab-
acute pain resolves within weeks of the initial causative insult, whereas olism and therefore influence the concentration of a drug present in
chronic pain is continuous, long-term pain which can last months or circulation. The primary catalytic function of CYP enzymes is identified
years [13]. as the transfer of one oxygen atom from molecular oxygen into various
substrates. Additionally, some CYP enzymes act as isomerases, reduc-
Chronic pain tases, dehydrases, or nitric oxide (NO) synthases, and some can catalyse
oxidative cleavage of esters [27].
The IASP defines chronic pain as that pain which persists past the CYPs are responsible for approximately 80% of all phase I metabo-
normal time of healing following an injury. However, since determining lism reactions (Fig. 1) [27,28]. Genetic changes in CYP genes can result
the end of the healing phase is problematic, most clinical definitions use in alterations in enzyme activities. One CYP isoform which has been ex-
a fixed time of persistent pain after initial onset. This demarcation be- tensively studied with respect to genetic variation is CYP2D6. This en-
tween acute and chronic pain is somewhat arbitrary, and typically zyme accounts for a very small percentage (b 2%) (Fig. 1b) of all CYPs
ranges from three months (e.g., for post-herpetic neuralgia) to six expressed in the human body, but is responsible for the metabolism of
months (e.g., for chronic low back pain) [14,15]. The treatment of chron- almost 20% of all drugs (Fig. 1a), including many drugs used in manage-
ic pain can be difficult because it is a complex condition influenced by ment of pain [29]. Most of these enzymes are polymorphic [30]; more
genetic makeup as well as physiological and psychological factors. than 80 CYP2D6 variants have been identified which, functionally, are
Chronic pain is a problem in many societies, with prevalence ranging associated with four general phenotypes: extensive metabolizer (EM),
from 2% to 40% in the adult population, resulting in social and economic intermediate metabolizer (IM), poor metabolizer (PM) and ultra-rapid
impacts [16–18]. Chronic pain can interfere with an individual’s activi- metabolizer (UM) [31]. The prevalence of these phenotypes varies by
ties of daily living, leading to work or school absenteeism, inability to ethnicity [22] and there are several examples in which phenotypic dif-
participate in leisure activities, sleep disturbances and problems eating. ferences in individuals have led to adverse drug reactions.
In the United States, one in three Americans experiences chronic pain In 2004, a case reported by Gasche and colleagues described a 62-
and chronic pain is the most common reason for people to visit their year-old male, with a history of chronic lymphocytic leukemia, who
family doctor [3]; one in five Canadians is reported to suffer from presented with fatigue, dyspnea, fever and cough with a bronchoalveo-
chronic pain [2]. It is therefore important that chronic pain be diagnosed lar lavage culture revealing yeast [33]. He was treated with codeine as a
accurately and treated effectively. Recently, an increased focus on pain cough suppressant. However on day 4 of treatment, he deteriorated rap-
management has led to a six-fold increase in the sales of prescription idly and became unresponsive. He was given naloxone, an opioid recep-
opioids in the U.S. between 1997 and 2006 [19]. Use of analgesics has in- tor antagonist, which resulted in rapid improvement in his condition.
creased from 7.2% of the U.S. population in 1988–1994 to 10.2% during Several factors were at play in this case, leading to the patient’s rapid de-
the period 2007–2010 [20]. terioration. Genetic studies revealed that he had CYP2D6 duplications,
causing him to be an ultra-rapid metabolizer of the pro-drug (codeine)
Pharmacogenetics and pain management to the active metabolite (morphine). Furthermore, CYP3A4 is also in-
volved in codeine metabolism, specifically in the conversion of codeine
Pharmacogenetics refers to the way in which genetic differences to (relatively inactive) norcodeine. In this case, co-treatment with
between individuals influence patient drug responses and drug disposi- clarithromycin and voriconazole, both CYP3A4 inhibitors, resulted in
tion [21]. Empirically, it is well understood that large inter-individual decreased conversion of codeine to norcodeine, shunting more codeine
variations exist with respect to the response to analgesics [22].With into the CYP2D6 morphine pathway. The patient also developed acute
conventional drug dosing, some patients will experience toxicity renal failure which resulted in the accumulation of glucuronide metab-
whereas other patients will not receive adequate analgesia from the olites with opioid activity (morphine-6-glucuronide) [33].
same dose. Variations in drug efficacy can vary as much as 2- to 10- Opioids, including codeine, are among the pain medications metab-
fold or even 100-fold among members of the same family [23–25]. olized by CYP2D6. In 2006, Koren and colleagues reported a case of a
These differences can be due to pharmacodynamics factors, based on breast-fed infant who died at 13 days of age [34]. The infant’s mother
variations in drug target receptors and downstream signal transduction, had been prescribed codeine for episiotomy pain. Stored breast
and pharmacokinetic factors, which affect drug metabolism and/or milk was found to contain higher than expected levels of morphine
elimination, altering the relationship between drug dose and steady (87 ng/mL; expected 1.9–20.5 ng/mL). CYP2D6 genotyping revealed
state serum drug concentrations. Development of tolerance, which that the infant’s mother was heterozygous for a CYP2D6 duplication,
may occur through both dynamic and kinetic mechanisms, can also making her an UM of codeine to morphine. The infant died from CNS
play a significant role in this response variation. depression and this, along with the genetic results, were consistent
Genetic variability is one factor playing a role in the way in which with opioid toxicity resulting from increased conversion of codeine to
drugs affect physiology. Generally, genes affecting outcome of treatment morphine. This case prompted the FDA in the USA as well as Health

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
4 B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx

a) b)

Fig. 1. Cytochrome p450: Abundance and proportions. a) Proportions of drugs metabolized by CYP 450 enzymes. Adapted from: Rendic S and Di Carlo FJ [27]. b) CYP Abundance in
human liver. Adapted from Yeo et al. [32].

Canada to advise physicians against the prescription of codeine to One study, which examined individuals prescribed oxycodone,
nursing mothers [35,36]. found that people homozygous or heterozygous for variant ABCB1/
Studies have also shown that CYP polymorphisms have an influence MDR1 alleles reported larger decreases in pain, but also a higher
on the metabolism of non-steroidal anti-inflammatory drugs (NSAIDs) frequency of adverse drug reactions to oxycodone, presumably due to
and have the potential to cause adverse drug reactions, most notably decreased efflux and therefore higher plasma concentrations of the
gastrointestinal (GI) bleeding, a frequent reaction resulting in drug [42]. Genetic variability of ABCB1/MDR1 has also been found to
significant patient morbidity and mortality. The risk of GI bleeding is be associated with inter-individual differences in pain relief achieved
particularly high with higher doses of NSAIDs [37]. Many NSAIDs are by morphine [43]. In a study of Korean patients receiving intravenous
metabolized by CYP2C9, which has been shown to have two allelic var- fentanyl, certain ABCB1/MDR1 genotypes were associated with
iants, referred to as CYP2C9*2 and CYP2C9*3 [37]. The CYP2C9 enzymes prolonged fentanyl-induced suppression of respiration rate [45].
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 metab- Cyclooxygenases 1 and 2
olism, and thus prolonged action, of NSAIDs in individuals with either of
these two alleles [38,39]. Cyclooxygenase 1 and 2 are encoded by two separate genes, PTGS1
Due to their platelet-inhibiting action, NSAIDs are also responsible (COX1) and PTGS2 (COX2). Genetic variation in either of these enzymes
for an increased risk of bleeding, particularly in patients taking the anti- would be expected to cause altered pharmacodynamic responses to
coagulant warfarin [38], since CYP2C9 is also important for warfarin NSAIDs. In a study which investigated post-surgical expression of
metabolism. Patients with the CYP2C9 *2 or *3 genotypes taking both PTGS1 and PTGS2, in relation to SNPs in each gene, certain SNPs were
an NSAID and warfarin have been shown to have a significantly higher shown to be associated with altered expression of the PTGS2 transcript
risk for an elevated prothrombin time, compared to patients with the [46]. The authors also reported differences in patient response to two
wild-type 2C9 genotype [38]. NSAIDs, rofecoxib, a selective COX-2 inhibitor and ibuprofen, a non-
Celecoxib is a selective COX-2 inhibitor which is metabolized by selective COX inhibitor. In particular, the G → C polymorphism at
CYP2C9. In a study of patients prescribed celecoxib for rheumatoid or − 765, in the promoter region of PTGS2, was found to be associated
osteoarthritis, patients with the *2 or *3 genotypes showed an increased with lower PTGS2 expression in individuals heterozygous (G/C) or ho-
elimination half-life compared to patients with the wild-type genotype mozygous (C/C) for the minor allele, compared to those homozygous
[40]. In a similar study, the investigators looked at patients prescribed for the major allele (G/G) [46]. Subjects with G/G genotype also
one of several different NSAIDs, including celecoxib, ibuprofen and reported lower pain intensity with rofecoxib treatment for 48 h follow-
naproxen. They grouped the patients into those who experienced GI ing surgery, compared to G/C or C/C subjects. This difference was not
bleeding and those who did not, and performed CYP 2C9 genotype anal- demonstrated for patients treated with ibuprofen. However, subjects
ysis on all patients. Within the group of patients with GI bleeding, a with G/C and C/C genotypes reported decreased pain intensity with ibu-
higher number of individuals had the *2 or *3 genotype compared to profen treatment at 48 h following surgery, compared to G/G patients.
the group not experiencing gastrointestinal bleeding [41]. The increased This difference was not seen in subjects treated with rofecoxib [46].
risk of bleeding was believed to result from reduced CYP2C9 enzyme ac- These data suggest that patients with increased PTGS2 expression
tivity in patients with the *2 or *3 alleles, leading to increased NSAID (G/G genotype) benefitted more from treatment with rofecoxib, where-
concentrations [41]. as patients with decreased PTGS2 expression (G/C or C/C genotypes)
benefited more from treatment with ibuprofen [46]. These findings
P-glycoprotein are likely explained by the selectivity of rofecoxib for COX-2, encoded
by PTGS2.
The ABCB1/MDR1 gene encodes P-glycoprotein, an efflux transporter
that influences drug transport in several tissues, particularly the Opioid receptor μ
intestine, kidney and brain [42,43]. P-glycoprotein is highly expressed
in endothelial cells of the brain vasculature, and is believed to effect Opioid receptor μ is the primary site of action for many endogenous
the efflux of drugs across the blood brain barrier [43]. ABCB1/MDR1 is opioids, as well as those used for analgesia. A number of SNPs have been
a highly polymorphic gene, with 38 single nucleotide polymorphisms described in OPRM1, the gene encoding this receptor. The most well
(SNPs) identified in the coding region [44]. The activity of P- characterized SNP in OPRM1 is A118G. A study by Janicki et al. [47]
glycoprotein is thought to be affected by genetic variability, with certain found the minor allele (G) present at lower frequency in subjects with
SNPs leading to decreased P-glycoprotein expression and activity. chronic pain treated with opioids, compared to a control group of opioid

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx 5

naïve patients, suggesting that the G allele may confer protection Clinical use of pharmacogenetics in pain management
against pain.
Epidural opioids are commonly used to provide analgesia to women Ideally, pharmacogenetic studies aim to aid in the selection and
during labor. Women heterozygous or homozygous for the OPRM1 dosing of an optimal drug therapy for a specific patient. Choosing the
A118G allele have been demonstrated to have higher pressure pain optimal therapy should lead to maximized therapeutic benefit,
thresholds than women homozygous for the more common A allele improved patient adherence and a reduction in adverse drug reac-
[48]. The A → G nucleotide substitution leads to an amino acid change tions [55]. The cases of opioid overdose discussed above illustrate in-
from asparagine to aspartic acid, and is thought to result in higher bind- stances where knowledge of patient genotypes may have been useful
ing affinity of β-endorphin to the opioid-μ receptor. In another study of to improve patient outcomes. However, most pharmacogenetic studies
women in labor, the median effective dose (ED50) of intrathecal fentanyl to date have examined variability in single candidate genes, such as
required to achieve effective analgesia was significantly higher in CYP2D6, and associated outcomes [22]. There are limitations to this ap-
women homozygous for the A allele (26.8 μg) compared to women proach, as very few drugs are metabolized by a single enzyme. Genome-
heterozygous or homozygous for the G allele (17.7 μg) [49]. wide association studies (GWAS) may possibly improve upon this limi-
tation. Furthermore, in addition to genetics, many other factors affect
drug responses, such as patient age, disease co-morbidities and, in
Catechol-O-methyltransferase (COMT) particular, co-medication [22,56] (Fig. 2). Genetics can only partially
explain the variability in patient responses to analgesic drugs. Patients
The COMT enzyme is responsible for the inactivation of catechol- are very commonly prescribed several medications for multiple
amines such as dopamine, norepinephrine and epinephrine. Several co-morbidities. Some medications induce CYP enzyme activity [57], as
polymorphisms have been identified in the COMT gene; however, the illustrated in the case above [33], while others inhibit activity [57],
most widely studied variant is a G to A nucleotide substitution resulting which can lead to altered metabolism unrelated to the patient’s CYP
in an amino acid change from valine to methionine at codon 158 phenotype. In general, pharmacogenetic studies thus far in pain man-
(Val158Met). This amino acid change is reported to decrease the agement have failed to yield evidence of improved clinical outcomes as-
thermal stability of the COMT protein, resulting in reduced enzymatic sociated with knowledge of patient genotypes when prescribing pain
activity. In studies of cancer patients experiencing chronic pain, patients medications. As GWAS studies continue, and panels of gene testing be-
homozygous for the more common Val 158 were found to require come more widely accessible, pharmacogenetics of pain management
higher doses of morphine to achieve analgesia, compared to patients may yet become more clinically useful [58].
heterozygous or homozygous for Met 158 [50,51]. These findings
suggest that variable COMT activity may influence the effects of opioids.
Animal studies have demonstrated reduced neuronal enkephalin con- Genetics and pain susceptibility
tent in the brain with chronic activation of dopaminergic transmission.
The reduced enkephalin content has been shown to be followed by an An individual’s genetic susceptibility to chronic pain is thought to be
upregulation of the opioid-μ receptor [52,53]. The lower doses of mor- complex, involving multiple genes, similar to the genetics of other
phine required by individuals with the Met158 variant may be ex- chronic diseases, such as diabetes [21]. As discussed above, several can-
plained by reduced COMT activity which is associated with increased didate genes have been identified, with specific mutations shown to
dopaminergic stimulation, resulting in upregulation of opioid-μ alter pain perception profiles between individuals. Examples include:
receptor expression in the brain, making morphine more effective. OPRM1, the gene encoding the μ-opioid receptor; COMT, encoding
Another study demonstrated differences in morphine side effects, catechol-O-methyltransferase, involved in the metabolism of catechol-
such as drowsiness, confusion and hallucinations, associated with amines; GCH1, encoding an enzyme involved in phenylalanine metabo-
certain COMT variants, which influenced how well patients tolerated lism and the production of dopamine; the melanocortin-1 receptor with
morphine [54]. mutations showing sex-specific differences in pain perception; and

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

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
6 B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx

members of the cytochrome P450 enzyme family, important for the metabolic acidosis and respiratory depression. More recently, cardio-
biotransformation of many drugs. Genome-wide association studies vascular risks associated with the use of NSAIDs have gained attention.
and other types of genetic investigations will likely identify many NSAIDs, particularly COX-2 selective agents, such as celecoxib, have
more genes involved in pain perception. been reported to increase the risk for thrombotic events, myocardial in-
farction and stroke in patients with coronary artery disease and athero-
sclerosis [68,69]. These findings have prompted the American Heart
Chronic pain management
Association to recommend use of acetaminophen or non-acetylated sa-
licylates for the treatment of chronic pain in patients with coronary ar-
Successful pain management can be viewed as providing adequate
tery disease [70]. Although classified with nonsteroidal anti-
analgesia without excessive adverse effects [59]. In 1982, Rane and col-
inflammatory agents, acetaminophen has minimal peripheral anti-
leagues [60] proposed a pharmacological approach to treat cancer pain
inflammatory activity.
with morphine. They proposed a step-wise approach that was later
IV acetaminophen has become a part of the multimodal analgesia
adopted by the World Health Organization (WHO) in 1986 [61,62]. Al-
approach for pain. Intravenous acetaminophen was made available
though this WHO stepladder approach was originally aimed at the
over 10 years ago in Europe, but in the United States only very recently.
treatment of chronic cancer pain (Fig. 3) [61,62], it is also widely used
Three randomized placebo controlled trials provided the bases of its ap-
in the treatment of chronic non-cancer pain [63]. The WHO analgesic
proval. Clinical studies show that for management of pain, intravenous
ladder recommends initial treatment of pain with non-opioids, such as
acetaminophen injection was at least as effective as morphine injection
NSAIDs and acetaminophen. If pain persists, treatment with a weak opi-
in renal colic and oral ibuprofen after cesarean delivery. In children a
oid, such as codeine or tramadol is recommended, followed by a strong
single-dose acetaminophen injection was similar to meperidine
opioid, such as morphine, until the patient is free of pain. At each stage
intramuscular (IM) for pain after tonsillectomy [71]. Effects begin 5 to
of the treatment ladder, adjuvant medications, such as antidepressants
10 min after IV administration, and peak effects occur in about 1 h
or anticonvulsants, may also be given to aid in alleviating patient anxi-
with an effective duration of 4 to 6 h. Table 1 provides pharmacokinetic
ety; some of these adjuvant drugs may also act directly to counter
details of specific NSAIDs and acetaminophen, as used in pain
pain [4,61]. Evident from this scheme is the fact that opioids remain
management.
the mainstay of chronic pain management and will be discussed further
below.
Opioids and pain management (Table 2)
Opioids remain the most potent analgesics available and are the
Drugs used in pain management mainstay of chronic pain management in both cancer patients and pa-
tients with non-malignant pain [2,72]. To manage chronic cancer pain,
NSAIDs and acetaminophen in pain management (Table 1) oral morphine is commonly administered at regular intervals. Chronic
NSAIDs such as aspirin and ibuprofen, as well as acetaminophen, act dosing can lead to pharmacological tolerance, but not to therapeutic
by inhibiting the enzymes cyclooxygenase-1 and -2, which catalyze the failure. In simple pharmacological terms, opioid-induced tolerance can
synthesis of prostaglandins. The inhibition of prostaglandin synthesis be described as a “shift to the right” in the dose–response curve; that
results in the analgesic, anti-pyretic and anti-inflammatory properties is, a higher dose is required over time to maintain the same level of an-
of these drugs, with the exception of acetaminophen, which does not algesia. Different mechanisms have been proposed for the development
show anti-inflammatory affects. Due to the widespread use of NSAIDs, of tolerance. Säwe and colleagues [73] investigated the metabolic corre-
they are the drugs most commonly associated with adverse effects, late to the development of tolerance to therapeutic effects by measuring
the most common of which are those involving the GI tract. Reduced morphine, morphine-3- and morphine-6-glucuronide in plasma and
prostaglandin levels in the gastrointestinal mucosa decrease mucus urine samples. They showed that the conjugation of morphine with glu-
and bicarbonate secretions, thereby reducing their protective effects curonic acid is proportional to the dose during long-term treatment
against the acidic gastric environment. In addition, NSAIDs can be di- with escalating doses, suggesting that this metabolic pathway is subject
rectly toxic to the gastric mucosa, leading to ulceration, and potentially to neither auto-induction nor saturation. Both in vitro and in vivo studies
fatal bleeding in the most extreme cases. Furthermore, reduced prosta- have shown that, on the cellular level, chronic opioid treatment leads to
glandin synthesis in the kidney has been shown to cause renal impair- a rapid reduction of agonist response, accompanied by internalization
ment. Chronic or acutely toxic acetaminophen exposure can lead to of opioid receptors [74]. These early adaptive processes as well as
liver toxicity through hepatocellular necrosis [64,65]. Additionally, aspi- long-term adaptations, such as receptor downregulation, or counter-
rin (acetylsalicylic acid) is associated with Reye’s syndrome, primarily regulatory processes such as adenylate cyclase superactivation, have
affecting children [66,67], and high doses of salicylates can cause been suggested to be crucial to the development of opioid tolerance
[75–77].
In recent years, there has been an increased focus on pain manage-
ment which has resulted in a societal escalation in opioid use, with opi-
oids 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 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].
The analgesic properties of opioids begin with their binding to opioid
receptors in the central nervous 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 them-
selves can be classified, based on their interactions with these receptors,
Fig. 3. The WHO three-step analgesic ladder. Adapted from [60,61]. Legend: The pain lad- as agonists, mixed agonists–antagonists and antagonists. The binding of
der 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
opioids to their G-protein coupled receptors produces signals causing
“steady state” or full effect is achieved. *Adjuvants: antidepressants, anticonvulsants, hyperpolarization of neuronal cell membranes and the suppression of
steroids. neurotransmitter release, resulting in analgesia [78,80,81].

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx 7

Table 1
Properties of NSAIDs and acetaminophen used in the management of chronic pain⁎.

Drug# Common route of Protein binding Bioavailability# Route of elimination Vd t½


administration L/kg (h)

Acetylsalicylic acid Oral 99% 80%–100% Renal 0.1–0.2 2–3


(aspirin ®) [164] (low doses)

Ibuprofen [165] Oral 99% 49%–73% Renal 0.14 0.9–2.5

Indomethacin [166] Oral 98% ~100 %(oral), Renal 0.4–1.2 5–10


80% to 90% (rectal) Fecal

Acetaminophen [167] Oral 20% ~100% Renal 0.25 1–4

Celecoxib [168] Oral 97% unknown Renal 5.7–7.0 11–16

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

Issues with opioids Newer opioid formulations have been designed in efforts to prevent
The increased use of opioids for pain management has led to an ex- abuse. These include Embeda™, an extended-release morphine co-
pansion in opioid misuse, resulting in an increased number of emergen- formulated with the opioid antagonist naltrexone, produced by Pfizer;
cy room visits related to drug-seeking behavior as well as in the number and an extended-release form of oxycodone, called OxyNeo™, by
of opioid-related overdose deaths [82,83]. Although opioids are among Purdue Pharma [84]. OxyNeo™ contains polyethylene oxide, which
the most commonly used analgesics, their clinical use is limited by the forms a viscous gel when in contact with water, rendering it unsuitable
development of tolerance and physical dependence. Another growing for injection or snorting, and also more difficult to chew or crush [87].
concern relates to the diversion of opioids from patients to other Buprenorphine, a semi-synthetic opioid was approved by the FDA
individuals not under medical supervision. in 2002. Sublingual preparations, used in addiction medicine, are
Opioids have several well-known side effects, including nausea, seda- co-formulated with the antagonist naloxone to prevent abuse.
tion and bowel dysfunction [84]. The most serious potential adverse effect
is respiratory depression and failure, which is the most common cause of
Specific opioids used in pain management
death related to opioid use [80]. Prolonged use leads to tolerance,
Opioids can be classified as either weak or strong, depending on their
resulting in higher dose requirements, which can lead to physical depen-
“analgesic ceiling”. This term is defined as the dose above which a drug
dence, demonstrated by withdrawal symptoms in patients experiencing
has no further effect on pain [88]. Weak opioids include tramadol, codeine
chills, muscle aches, vomiting, diarrhea, hyperthermia, anxiety and hostil-
and hydrocodone; strong opioids include morphine, methadone,
ity [80]. Additionally, opioid use can result in feelings of euphoria which
buprenorphine, fentanyl, oxycodone, hydromorphone, oxymorphone,
can promote compulsive use leading to psychological dependence.
meperidine and levorphanol. Detailed pharmacokinetic information for
A detailed medical assessment is recommended prior to prescription
each of these drugs can be found in Table 2.
of opioids. This should include a detailed medical history, review of
medical records, urine toxicology screen and psychological evaluation,
including questionnaires aimed at determining an individual’s risk for Weak opioids
abuse [84,85]. This is of particular importance in those populations at
higher risk for opioid misuse. Chronic pain frequently occurs in individ- Codeine
uals with psychological conditions, including anxiety and depression; it
is estimated that 30%–60% of individuals with depression also experi- Codeine is considered a weak opioid, and is used for its analgesic,
ence chronic pain [86]. Psychological conditions are often comorbid in anti-tussive and anti-diarrheal properties. It is a pro-drug which re-
individuals with substance-abuse disorders, resulting in a high risk quires biotransformation via hepatic CYP2D6 to the active metabolite
for opioid abuse in many chronic pain patients including those with morphine [89]. Approximately 5%–10% of codeine is converted to mor-
substance abuse issues and mental health disorders [85]. phine in the liver, while the remainder is either conjugated, forming

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
Table 2

8
Properties of opioid medications commonly used in the management of chronic pain.
j.clinbiochem.2014.05.065
Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/

Drug# Common route of Protein binding Bioavailability# Route of elimination Vd⁎⁎ t½⁎⁎ CYP
administration⁎ L/kg (h)

Buprenorphine [170,171] TD, B, IV 96% 31% Renal 2–3 (IV)⁎ 2–4 (parental) 3A4
(sublingual) biliary 19 (B) 18–49 (sublingual)
26 (TD)

Codeine [89] Oral b1% ~90% Renal 2.5–3.5 1.2–3.9 3A4, 2D6
(oral)

B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx


Fentanyl [89,172] TD, oral 79% 92% (transdermal) Renal 3–8 3–12 3A4
89% (intranasal)
50% (buccal)
33% (ingestion)

Hydro Hydrocodone codone [89,90] Oral 20%–50% ??????? Renal 3.3–4.7 3.4–8.8 2D6, 3A4

Hydromorphone [89,173] Oral, IV 19% 30%–35%, (Oral) Renal 2–4 3–9 glucuronidation
52%–58%
(intranasal)

Levorphanol [89,120] IV, oral 40% 70% (oral); 100% (IV) Renal 10–13 11–16 glucuronidation

Methadone [108,109] Oral 87% 41%–99% Renal 4–7 15–55 3A4 N 2B6 N 2D6
(oral)
Morphine [89,98] IV, IM, oral 35% 20%–40% Renal (90%) 2–5 1.3–6.7 Glucuronidation (major)
j.clinbiochem.2014.05.065
Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/

(oral), Biliary (10%) 2D6 (minor)


36%–71% (rectally), 100%
(IV/IM)

Meperidine (Pethidine) [174,175] Oral, IV, IM 45%–64% 50%–60% (Oral), hepatic 3.7–4.2 2–5 2B6, 3A4 & 2C19(minor)
80%–90%
(Oral, in cases of hepatic impairment)

Oxycodone [114] Oral 45% 60%–87% Renal 1.8–3.7 3–6 2D6

B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx


Oxymorphone [115] Oral 10–12% 10% Renal 2.4 4–12 glucuronidation
(oral), fecal
40% (Intranasal), 100%
(IV, IM)

Remifentanyl i.v 70% (bound to plasma proteins) Not applicable - - 1–20 min Nonspecific esterases
Given iv

Tramadol [89,95] Oral 15–20% 70%–75% Renal 2.6–2.9 4.3–6.7 2D6 N 2B6, 3A4
(oral),
77%
(rectal),
100% (IM)

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

9
10 B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx

codeine-6-glucuronide (active), or methylated via CYP3A4, producing system [89,100,101]. Given intravenously, fentanyl has a very rapid
norcodeine (inactive) [89]. onset but short duration of action, and is thus administered as a contin-
uous infusion. In managing pain, fentanyl is most often provided
Hydrocodone through transdermal patches or buccal tablets [89]. This allows the
drug to distribute throughout fatty tissues, leading to slower release
This is a semi-synthetic drug, similar to codeine in structure and to and prolonged effects. Fentanyl undergoes extensive pulmonary
morphine in its effects [89]. Hydrocodone is metabolized by CYP2D6 first-pass metabolism as well as hepatic metabolism to the inactive
to active hydromorphone, which binds to the μ-opioid receptor, and metabolite norfentanyl via CYP3A4 [89].
by CYP3A4 to the inactive metabolite norhydrocodone [90].
Remifentanil
Tramadol
Remifentanil is a μ-opioid receptor agonist with an analgesic potency
Tramadol is a synthetic codeine analog that binds to the μ-opioid similar to that of fentanyl. It was studied for analgesic efficacy correlat-
receptor and inhibits the reuptake of serotonin and norepinephrine ing with expression of the serotonin transporter (5-HTT), as serotonin
[89]. Tramadol has a lower risk of addiction than other opioids; howev- can influence the antinociceptive effects of opioids at the spinal cord.
er, it is associated with two significant adverse drug reactions: seizure Remifentanil has a significantly better analgesic effect in individuals
and serotonin syndrome [91]. Tramadol seizures usually occur in pa- with a genotype coding for low 5-HTT expression (SA/SA and SA/LG)
tients already taking anticonvulsant medications. Serotonin syndrome compared to those with high expression (LA/LA) [102]. It is predomi-
is characterized by neuromuscular and autonomic hyperactivity as nantly metabolized by non-specific esterases [103], and because of its
well as altered mental status, due to excess serotonin activity in the rapid systemic elimination and ultra-short half-file, it has a pharmacoki-
CNS. Higher risk of this syndrome is associated with tramadol overdose netic advantage in clinical situations requiring predictable termination
or co-administration of antidepressant medications, such as tricyclic of effect such as analgesia during labor [104]. In a systematic review,
antidepressants and selective serotonin reuptake inhibitors (SSRIs), Leong and colleagues compared remifentanil with meperidine for
which, in addition to tramadol, inhibit serotonin reuptake, leading to labor analgesia. They found remifentanil to be superior in reducing
high serotonin levels in the synaptic space[92,93]. mean visual analog scale pain scores for labor pain after 1 h [105].
Tramadol is metabolized by CYP2D6 in the liver to its main metabo- Remifentanil crosses the placenta, but is rapidly metabolized and
lite, O-desmethyl-tramadol which shows a higher affinity for opioid re- redistributed. Although maternal sedation and respiratory changes do
ceptors than the parent drug [94]. Metabolism to the minor metabolite, occur, they are without adverse neonatal or maternal effects [106].
N-desmethyl-tramadol occurs via CYP2B6 and CYP3A4 [95].

Strong opioids Methadone

Morphine Methadone is a synthetic opioid with morphine-like effects. It is com-


monly used in the treatment of opioid addiction, but also in the treat-
Morphine increases the threshold for pain perception by binding ment of chronic pain [89,107,108]. Given orally, methadone has a
strongly to the μ-opioid receptor; it also has activity towards κ- and much higher bioavailability compared with oral morphine (70%–90%,
δ-opioid receptors. Morphine administered orally undergoes a large compared to 38%, respectively). Due to its relatively slow metabolism
first-pass effect, resulting in oral bioavailability of 38% on average [96]. and high lipid solubility, methadone also has a long half-life (between
The major morphine metabolites result from glucuronidation by 8 and 60 h), leading to longer term analgesia than with morphine [107,
the hepatic isoenzyme UGT2B7 to inactive morphine-3-glucuronide 108]. Methadone is extensively metabolized by CYP3A4, and to a lesser
(approximately 60%) and active morphine-6-glucuronide (up to 10%) extent by CYP1A2, -2D6, -2D8, -2C9/2C8, -2C19, and -2B6, to its inactive
[97]. CYP2D6 plays a minor role in morphine metabolism. Side effects metabolite, 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP)
of morphine include sedation, nausea, a feeling of warmth, urinary [107–110]. Caution must be exercised when co-prescribing methadone
retention, euphoria, reduced ability to concentrate and constipation. with QT-prolonging cardiac drugs due to methadone’s ability to interact
The most serious side effect of morphine is potentially fatal respiratory with voltage-gated potassium channels in the myocardium, also leading
depression with morphine toxicity [89,98]. to QT prolongation [111] and ventricular arrhythmias such as Torsades
de Pointes. Significant drug–drug interactions have been reported for
Hydromorphone methadone, when combined, for example, with fluoxetine, quetiapine,
or promethazine [110].
Hydromorphone, a hydrogenated ketone of morphine, is a synthetic
opioid with effects similar to morphine [89]. When given orally, Buprenorphine
hydromorphone is rapidly absorbed by the GI tract and undergoes
extensive first-pass metabolism in the liver. Like morphine, it is As an alternative to methadone, buprenorphine can be used for the
glucuronidated in the liver to hydromorphone-3-glucuronide and treatment of opioid addiction [89]. It has also been approved for
hydromorphone-6-glucuronide; hydromorphone-3-glucuronide has the treatment of moderate to severe pain [112]. Similarly to morphine,
no analgesic properties, but has been shown to exhibit neuroexcitatory buprenorphine must be used with caution in the setting of co-
effects [99]. Hydromorphone suppresses the cough reflex, similar to administration of other QT-prolonging medications or in individuals
other opioids. Side effects of hydromorphone are dose-related and can with hyperkalemia [112]. Buprenorphine has a ceiling effect with
include mood changes, euphoria, nausea, vomiting, and hypotension, respect to respiratory depression, thereby reducing the likelihood of
as well as respiratory depression in large doses [89]. this potentially fatal consequence, making it an attractive choice for an-
algesia [113]. Given orally, buprenorphine undergoes extensive first-
Fentanyl pass metabolism and is therefore usually administered by transdermal
or buccal routes to improve bioavailability [112]. Prolonged analgesia
Fentanyl is a highly lipophilic, synthetic opioid which is 100 times can be achieved with buprenorphine due to its lipophilic properties
more potent than morphine. Its potency is due to its efficiency in cross- and its slow dissociation from opioid receptors. Buprenorphine is
ing the blood brain barrier, rapidly gaining access to the central nervous metabolized by CYP3A4 in the liver primarily to nor-buprenorphine.

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx 11

Oxycodone CYP2D6, to the potent metabolite oxymorphone (see below) as well


as noroxycodone, a weakly active metabolite [114].
Oxycodone is a synthetic opioid, similar in structure to codeine and
prescribed for moderate to severe pain [89]. This drug is commonly Oxymorphone
compounded with other drugs, such as acetaminophen or ibuprofen
[89]. Side effects of oxycodone treatment include euphoria, sedation, Oxymorphone offers excellent analgesia, comparable to that of
constipation, cough suppression and respiratory depression. The eupho- morphine, but with less sedative effects [115]. This results from the
ria caused by oxycodone is quite pronounced and can therefore lead to lipophilic nature of oxymorphone, allowing ease of access to the cen-
abuse of and dependence on the drug. Oxycodone is metabolized, via tral nervous system. Oxymorphone undergoes extensive hepatic

Table 3
Antidepressants commonly used in pain management.

Drug# Common route of Protein binding Bioavailability# Route of elimination Vd⁎ t½⁎ CYP
administration L/kg (h)

Amitriptyline [176] Oral N 90% 30%–60% Renal 6–10 8–51 2D6


due to first pass metabolism

Bupropion [177] Oral 85% ?????? Renal 40 4–24 2B6


Fecal

Citalopram [178] Oral 50% 80% Renal 12–16 25–40 2C19, 3A4, 2D6

Desipramine [179] Oral 70%–90% 73%–92% Renal 22–59 12–54 2D6

Duloxetine [180] Oral N90% 32% to 80% Renal 17–26 8–17 1A2,2D6
Fecal

Fluoxetine [181] Oral 94% 72% Renal 20–42 1–3 days 2D6
Fecal

Venlafaxine [182] Oral 27% 42% ± 15% renal 4–12 3–7 2D6, 3A4

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

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
12 B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx

first-pass metabolism, and its major route of metabolism is via is often a co-morbidity of chronic pain. Patients experiencing chronic
glucuronidation [89,115]. pain have been shown to have 2–5 times the risk of developing depres-
sion than the general population [122].
Meperidine (pethidine) Common TCAs used in pain management include amitriptyline and
imipramine (Table 3), administered at much lower doses for pain
Meperidine (Demerol™), also known as pethidine, is a synthetic management than for psychiatric indications [22]. These two drugs
μ-opioid receptor agonist that is structurally similar to atropine. are metabolized to the active metabolites nortriptyline and desipra-
Pharmacologically, it represents both morphine and atropine. This mine, respectively, by hepatic CYP2D6 [22]. Side effects of TCAs that
unique property of an analgesic with spasmolytic and sedative action can limit their use include weight gain, anticholinergic effects, hypoten-
accounts for its wide range of clinical applications [116]. Although sion and cardiovascular effects [22,121]. TCAs have been better charac-
meperidine efficacy has been questioned [117], a survey of intrapartum terized for their role in pain management than SSRIs [121].SSRIs used in
analgesia practice in United Kingdom and Norway reported that this pain management include fluoxetine and citalopram (Table 3). SSRIs are
opioid is still commonly used during labor [118,119]. often better tolerated than TCAs, due to their milder side effects; how-
ever, data regarding their efficacy in the treatment of chronic pain
Levorphanol have been inconsistent [121].
More recently, antidepressants able to target several neurotransmit-
Levorphanol is a synthetic opioid, similar in structure to morphine, ters, such as venlafaxine (serotonin, norepinephrine, and dopamine),
but more potent in its analgesic effects [89,120] and also has anticholin- duloxetine (serotonin and norepinephrine) and bupropion (serotonin
ergic effects [120]. This drug is usually given orally or intravenously and and dopamine) have been more widely used in pain management
is eliminated through glucuronidation in the liver [120]. (Table 3). Venlafaxine has been shown to be particularly efficacious
when prescribed in combination with gabapentin, a GABA analogue
Other medications in the treatment of chronic pain often used to treat neuropathic pain [123].

Antidepressants Anticonvulsants

Tricyclic antidepressants (TCAs) and selective serotonin re-uptake Anticonvulsants (Table 4) have been used in pain management since
inhibitors (SSRIs) are commonly used as adjuvant therapy in the the 1960’s when these drugs were originally introduced for the treat-
treatment of chronic pain. The mechanisms by which these medications ment of epileptic seizures [124]. These medications work by a variety
alleviate pain are not fully understood, but are believed to be mediated of mechanisms to suppress the rapid firing of neurons. Phenytoin, one
through their inhibition of neurotransmitter reuptake in the synaptic of the first anticonvulsants introduced, reduces neuronal excitability
cleft. These mechanisms are related to pain signaling in descending by blocking sodium channels. It has been used in the treatment of
spinal pain pathways [121]. In addition to their analgesic role, antide- chronic neuropathic pain, but today its use for this indication is limited
pressants are commonly used in pain management because depression due to side effects, namely, sedation and motor disturbances [125].

Table 4
Anticonvulsant drugs used in pain management.

Drug# Common route of Protein binding Bioavailability# Route of elimination Vd⁎ t½⁎ CYP
administration L/kg (h)

Carbamazepine [183,184] Oral 75% ?????? Renal 0.8–1.8 18–65 3A4

Gabapentin [185] Oral b 3% 27%–60% (inversely proportional to dose) Renal 0.8–1.3 5–9 Not metabolized

Lamotrigine [186] Oral 55% 98% Renal 0.9–1.3 12–62 glucuronidation

Phenytoin [187,188] Oral, IV 87%–93% 70%–100% Biliary 0.5–0.8 8–60 2C19


oral,
24.4%
(rectal, IV)

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

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx 13

Carbamazepine has largely replaced phenytoin in chronic pain neuralgia and diabetic neuropathy [124]. However, careful titration of
management and has been shown to be effective in the treatment of lamotrigine dose is required because of the risk of Stevens–Johnson
neuropathic pain, in particular trigeminal neuralgia [124,126] (a disor- syndrome, a life-threatening dermatological condition leading to cell
der characterized by episodes of intense facial pain, originating from necrosis, causing the epidermis to separate from the dermis [129].
the trigeminal nerve) [126]. Side effects of carbamazepine include
drowsiness, blurred vision, nausea and vomiting. There is a dose–effect
relationship and it can be difficult to achieve a therapeutic effect in pa- Triptans
tients who are sensitive to these side effects [125]. This is of particular
relevance in elderly patients, where conditions such as cardiac disease Triptans are serotonin agonists with high affinity for the 5HT1B and
and hypernatremia can further complicate adverse effects [127]; thus, 5HT1D receptors [130,131]. The serotonergic system is known to play a
close monitoring of these patients is required [124,125]. role in the pathophysiology of migraines, leading to the development
Newer anticonvulsants, such as lamotrigine and gabapentin, are and prescription of triptans for this condition. Binding of these drugs
showing promise in chronic pain management; gabapentin, in particu- to 5HT receptors in the brainstem and thalamus leads to the blockade
lar, has been shown to be well tolerated with few side effects [128]. of vasoactive peptide release from perivascular trigeminal neurons,
Gabapentin binds to voltage-gated calcium channels, leading to a reduc- and thus reduced cerebral vasodilation [131]. Currently, there are
tion in the release of the neurotransmitters glutamate and substance P. seven triptan molecules available, each with its unique pharmacokinetic
It has demonstrated effectiveness in the treatment of diabetic neuropa- and pharmacodynamic profile. The four most commonly prescribed and
thy, post-herpetic neuralgia, trigeminal neuralgia, multiple sclerosis, well characterized triptans are sumatriptan, naratriptan, zolmitriptan
migraine as well as in chronic pain caused by malignancy [128]. and rizatriptan (Table 5) [132]. Triptans are not recommended for mi-
Lamotrigine has been shown to be useful in the treatment of trigeminal graine treatment in individuals with poorly controlled hypertension,

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

Drug# Common route of Protein binding Bioavailability# Route of elimination Vd⁎⁎ t½⁎⁎ CYP
administration L/kg (h)

Almotriptan[131] Oral 35% 70% Renal 2.5–4 3–4 3A4, MAO⁎, 2D6
Fecal

Eletriptan[131] Oral 85% 50% Renal 2.1–3.3 3–7 3A4

Frovatriptan[131] Oral 15% 20%–30% Renal 3–4.2 20–30 1A2, 2D6

Naratriptan[131] Oral 20%–30% 74% Renal 2–3 5–6 Unknown CYP

Rizatriptan[131] Oral 14% 45% Renal 1.3–2.2 1.6–3.2 MAO, 1A2

Sumatriptan[131] Oral 14%–21% 15% Renal 1.3–4.6 1–4 MOA


(oral)
96%
(s.c)

Zolmitriptan[131] Oral, nasal 25% 40% (oral) Renal 7.0 1.6–3.8 1A2, MAO, 3A4
Fecal

#
All chemical formulas and bioavailability data are from their respective pages in Wikipedia.
⁎ MAO: monoamine oxidase.
⁎⁎ [169].

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
14 B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx

hepatic or renal impairment or coronary artery disease, as serotonergic now under consideration in different parts of the world including the
blockade may exacerbate these conditions [131]. United States. Synthetic cannabinoids, such as dronabinol and nabilone,
are available in the U.S. and Canada for use as antiemetics during cancer
Benzodiazepines chemotherapy, and as adjunct therapies for neuropathic and chronic
pain. They are not yet widely used, and are under further study to delin-
Benzodiazepines play a role in the treatment of insomnia, muscle eate their efficacy and pharmacological parameters in various clinical
tension, and, in particular, anxiety related to chronic pain [133]. Chronic contexts [143]. Side effects include increased heart rate, blood pressure
pain patients experience increased anxiety compared to the general changes, anxiety, psychomotor retardation, impaired memory and psy-
population, and this can often worsen the subjective experience of chosis. Cannabinoids are contraindicated in pregnancy, uncontrolled
pain. Evidence suggests that benzodiazepines are only effective in hypertension, active ischemic heart disease, arrhythmias and schizo-
treating acute anxiety related to chronic pain, but are not effective in phrenia. Clark and co-workers suggest guidelines for the use of these
the setting of chronic anxiety [133] (for which antidepressants have cannabinoid compounds [144].
been found to be more effective). The caveats to benzodiazepine treat- Cannabis and cannabinoids have a potential for dependence, which
ment for chronic pain are the potential for abuse as well as side effects, can lead to preoccupation and compulsion. Prescription of cannabinoids
including paradoxical reactions (increased hostility, psychosis and for chronic pain management requires clinician to appropriately select
behavioral disturbances) [133]. patient and adequately monitor to avoid the development of depen-
dence [134,136]. Recently, Donoghue and colleagues report the interac-
Cannabinoids tion effect of cannabis use on gender and age of onset of schizophrenia
and schizoaffective disorder. They show that use of cannabis is associat-
Cannabis is an annual plant that grows wild in regions of mild or ed with an earlier age of onset of schizophrenia. There is a significant
tropical weather, and has been used to treat pain for centuries. It is by interaction between gender and cannabis use whereby the gender
far the most widely cultivated, trafficked and abused illicit drug; half difference in age of onset is diminished in cannabis users [145].
of all drug seizures worldwide are of cannabis. The geographical spread
of those seizures is also global, covering practically every country of the Ziconotide
world. Delta-9-tetrahydrocannabinol (THC) is the active component
in cannabis, responsible for its analgesic and psychoactive effects. Ziconotide is a synthetic conopeptide that selectively inhibits N-type
Ingestion of THC results in feelings of euphoria, relaxation and a general voltage-gated calcium channels, reducing the release of pain-
sense of well-being. However, heavy cannabis use can result in halluci- modulating neurotransmitters. Conopeptides are derived from the
nations, anxiety and depression [134,135]. venom of predatory cone snails, and ziconotide is specifically derived
Endogenous cannabinoids, referred to as endocannabinoids, have from the venom of a Pacific fish-hunting snail, Conus magus [146].
been identified, as well as two endocannabinoid receptors, the G- Ziconotide has been approved for intrathecal administration by
protein coupled receptors CB1 and CB2. High levels of CB1 are expressed continuous infusion for the treatment of severe chronic pain in situa-
in the brain and spinal cord, particularly in areas known to be involved tions where patients are not responding to conventional therapies
in nociception. CB2 is not as well characterized, but has been shown to [147]. Evidence indicates that this drug is useful in the treatment of
have lower expression levels in the brain than CB1. Endocannabinoids both neuropathic and non-neuropathic pain. Intrathecal administration
are bioactive lipids which regulate neural activity by binding to CB1 is required because, given intravenously, ziconotide demonstrates poor
and CB2, causing inhibition of neurotransmitter release. Endogenous penetration of the blood brain barrier and results in sympatholytic
and exogenous cannabinoid activity results in anti-nociceptive effects effects leading to orthostatic hypotension [147]; oral preparations are
as well as short-term memory impairment, stimulation of appetite not available. Ziconotide is highly hydrophilic and is thought to move
and antiemetic effects [134–136]. slowly through the CSF to its target receptor, leading to a lag time to
THC acts synergistically with other analgesic agents, such as analgesia upon administration. The efficacy of ziconotide shows a
morphine and some NSAIDs, though the mechanism for the synergistic wide inter-individual variation [147,148].
action between morphine and THC is unknown [134]. In the case of The most common side effects of ziconotide include dizziness,
NSAIDs, cannabinoids and prostaglandin share a similar structure confusion, ataxia, memory impairment and hallucinations. However,
which is believed to result in a convergence of signals. Activity of CB1 re- there is also large inter-individual variation in the experience of side ef-
ceptors is increased by NSAIDs including ibuprofen and indomethacin. fects [147]. Toxicity appears to be related to the rate of infusion and is
Furthermore, CB1 receptor antagonists block the analgesic effects of reversible after discontinuation, but, because of low tissue diffusion,
ibuprofen [134]. the effects can persist for several weeks [147,148]. Data regarding
Evidence for the use of cannabinoids in pain management is lacking ziconotide metabolism are sparse. The drug is not believed to be metab-
due to the small number of clinical trials in this area. The best evidence olized in the CSF but rather cleared by transport into the systemic
for cannabinoid use in chronic pain comes from HIV and multiple sclerosis circulation where it is degraded by serum proteases [147].
studies in which cannabinoids were used to treat the chronic neuropathic
pain associated with these conditions [136]. Smoking cannabis 3–5 times Novel therapies in pain management
per week was shown to be effective in alleviating neuropathic pain
associated with HIV as well as AIDS-related anorexia [137,138]. Several Targeting epigenetic modifications
studies demonstrated benefit of oral treatment with synthetic THC for
central pain and spasticity associated with multiple sclerosis [139–142]. Epigenetics refers to functionally relevant genomic changes not in-
There are several issues associated with the use of cannabinoids in volving alterations in nucleotide sequence, in response to developmen-
pain management. Firstly, the method of drug delivery poses a problem. tal or environmental cues. Epigenetic mechanisms are manifested
Smoking of cannabis is the easiest delivery route and allows for ade- through dynamic, reversible chemical modifications in the genome
quate control of dose [135,136]. However, the adverse effects of and are involved in differential gene expression throughout life
smoking are well known, with THC contents varying depending on [149]. The most well characterized epigenetic modifications are DNA
where and how the cannabis plants are grown. Canada, which allows methylation and histone acetylation.
prescription of cannabis for chronic pain therapy, requires that patients DNA methylation is regulated by DNA methyltransferases, while his-
obtain their medicinal cannabis from a centralized source so that the tone modifications are largely regulated by histone deacetylases
THC content may be monitored over time [136]. Similar strategies are (HDAC) [149]. Abnormal DNA methylation [150] and histone

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx 15

acetylation [151,152] have been demonstrated to occur in pain states. It Therapeutic drug monitoring in pain management
has also been proposed that primary injuries may result in lasting epige-
netic marks leading to an increased risk of chronic pain [149], which In pain management, the rationale for drug monitoring has largely
may even persist trans-generationally. Drugs aimed at targeting the en- focused on monitoring patient adherence, detecting diversion, and
zymes responsible for epigenetic modifications are under development detecting the presence of illicit, non-prescription drugs. Traditionally,
and in some cases have been approved by the FDA. While these drugs drug monitoring in pain management patients has been in the form of
have been developed largely for the purpose of cancer treatment, they urine drug testing.
are also believed to have potential in pain management. In animal injury Urine drug testing has been demonstrated to reduce illicit drug
models, rat hind-paw injection of Freund’s complete adjuvant (CFA) use and has been widely used in monitoring patients in treatment
followed by intrathecal injection of zebularine, a DNA methyltransfer- for drug addiction [158,159]. However, most urine methodologies
ase inhibitor, was shown to reduce pain sensitivity [153]. In a similar have been adapted from occupational deterrent-based testing for
rat model, pre-injection treatment with HDAC inhibitors was reported illicit drug use and have not been optimized for use in the pain
to delay CFA-induced thermal hyperalgesia whereas post-injection management setting [160]. Given the chronicity of treatment drug
treatment was shown to reduce the intensity of CFA-induced thermal administration, and the wide range of drug half-lives (Tables 1–5)
hyperalgesia [154]. In human patients with type 2 diabetes, treatment a “positive” urine result is to be expected if a patient is adherent in
with valproic acid, which is a known to act as a strong the HDAC inhib- taking his/her medication. A “negative” urine results suggests non-
itor, conferred improved pain sensitivity scores after treatment compliance with the treatment protocol or urine sample tampering.
compared to before treatment [155]. 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
Gene therapy of the type of information urine drug testing can provide along
with an appreciation of the limitations of such testing [161].
Gene therapy involves the use of viral vectors where the viral ge- Urine drug concentrations are often calculated relative to urine
nome is replaced with nucleic acid sequences encoding a promoter to creatinine concentrations in order to correct for in vivo dilution, which
drive gene expression as well as a transgene of interest [156]. For pain can vary through time, depending on an individual’s fluid intake.
management therapies, the transgene encodes for an analgesic agent. However, these calculations assume stable renal function and creatinine
Gene therapy based approaches have several advantages in pain production, an assumption which can lead to errors if unjustified. Pain
management. In contrast to pharmacotherapies, gene therapy allows medications are usually prescribed to be taken in a chronic manner so
for persistent expression of a protein-based analgesic agent at the site that steady state plasma concentrations and therapeutic pharmacody-
of action. These methods avoid the first-pass effect and can improve namic effects can be achieved. Steady state drug concentrations in
bioavailability [156]. Gene therapy can also reduce side effects associat- serum imply that the urine excretion window (UEW) of the drug/me-
ed with systemic drug use. There are limitations to these approaches, tabolite is also at “steady state” in patients with stable renal function.
including inadequate transgene expression and the evocation of Fig. 4 shows the concentration of methadone metabolite 2-ethylidene-
immune responses against the viral vectors [156]. Nevertheless, new 1,5-dimethyl-3,3-diphenylpyrrolidine (EDDP) in a patient receiving
paradigms using non-viral insertion of therapeutic transgenes are methadone chronically. This figure clearly shows that urine creatinine
under way to bypass some of these limitations. correction does not add value to the EDDP adherence monitoring.
In a recent study, terminal cancer patients were given different gene Here, patient non-adherence can be detected when the urine EDDP
doses of human PENK, encoding preproenkephalin, the protein precur- concentration falls outside the UEW [162]. Extremely low creatinine
sor that is processed to 6-met-enkephalin and 1-leu-enkaphalin, endog- concentrations are indicative of potential adulteration and can be useful
enous opioid peptide ligands for the delta opioid receptor [157]. in detecting specimens that have been tampered with [161]. Unfortu-
Patients in the two highest virus dose groups reported decreases in nu- nately, urine drug testing is not effective in estimating serum drug
meric rating scale (NRS) pain scores of up to 50% from before treatment. concentrations or assessing drug efficacy.
Patients in the higher dose groups reported substantial reductions in In light of the limitations associated with urine drug testing, moni-
pain compared to patients in the lowest dose group. This study demon- toring of pain management patients through serum or plasma drug
strates proof-of-concept for gene therapy in pain management. measurements has been advocated. Therapeutic drug monitoring

Pt XY
5000 60.00
4500
50.00
4000
3500
UEW 40.00
EDDP ng/mL

pH and UCr

3000
2500 30.00
2000
20.00
1500 problem samples
1000
10.00
500
0 0.00

date of analysis
EDDP Creatinine PH

Fig. 4. EDDP urine excretion window at steady state. Legend: *UEW urinary excretion window. The green lines represent the 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine
(EDDP) excretion window.

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
16 B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx

(TDM) involves the measurement of serum or plasma drug levels at [3] Hardt J, Jacobsen C, Goldberg J, Nickel R, Buchwald D. Prevalence of chronic pain in
a representative sample in the united states. Pain Med 2008;9:803–12.
timed intervals to determine whether steady state drug concentrations [4] Organization W.H. Cancer pain relief with a guide to opioid availability; 1996.
are within the therapeutic or expected range given the drug dosage [5] Kroenke K, Krebs E, Wu J, Bair MJ, Damush T, Chumbler N, et al. Stepped care to op-
[163]. TDM can be used by clinicians to explain drug toxicity or lack of timize pain care effectiveness (scope) trial study design and sample characteristics.
Contemp Clin Trials 2013;34:270–81.
efficacy, and can aid in adjusting drug doses or in choosing specific [6] Treede RD, Jensen TS, Campbell JN, Cruccu G, Dostrovsky JO, Griffin JW, et al.
medications [163]. A major advantage of plasma drug measurement is Neuropathic pain: redefinition and a grading system for clinical and research
that it integrates many factors affecting drug metabolism, as outlined in purposes. Neurology 2008;70:1630–5.
[7] Gilron I, Watson CP, Cahill CM, Moulin DE. Neuropathic pain: a practical guide for
Fig. 2. Half-life measurement can be a functional marker of the cumulative the clinician. Can Med Assoc J 2006;175:265–75.
effect of pathophysiological and drug–drug interactions and provide an [8] Kroenke K, Krebs EE, Bair MJ. Pharmacotherapy of chronic pain: a synthesis of rec-
overall picture of the specific drug disposition and pharmacodynamics ommendations from systematic reviews. Gen Hosp Psychiatry 2009;31:206–19.
[9] Dubin AE, Patapoutian A. Nociceptors: the sensors of the pain pathway. J Clin Invest
in the individual patient. However, TDM requires the collection of serial
2010;120:3760–72.
blood samples from patients, which can be difficult and inconvenient [10] Tyrer S. Psychosomatic pain. Br J Psychiatry 2006;188:91–3.
[160,163]. Timing of sample collection also becomes an important consid- [11] Arendt-Nielsen L, Svensson P. Referred muscle pain: basic and clinical findings. Clin
eration, depending on whether peak or trough measurements (or both) J Pain 2001;17:11–9.
[12] Kooijman CM, Dijkstra PU, Geertzen JH, Elzinga A, van der Schans CP. Phantom pain
are to be monitored. The time window within which a particular drug and phantom sensations in upper limb amputees: an epidemiological study. Pain
or its metabolite can be detected in serum is an important consideration 2000;87:33–41.
for sample collection. To assist in appropriate clinical interpretation, [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
knowledge of tolerance and pharmacokinetics of particular drug(s) Res Manag 2011;16:445–50.
under investigation is essential. [15] Pergolizzi J, Ahlbeck K, Aldington D, Alon E, Coluzzi F, Dahan A, et al. The develop-
ment of chronic pain: physiological change necessitates a multidisciplinary
approach to treatment. Curr Med Res Opin 2013;29:1127–35.
Summary and conclusions [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.
The experience of chronic pain is one of the commonest reasons in- [17] Eriksen J, Ekholm O, Sjogren P, Rasmussen NK. Development of and recovery from
long-term pain. A 6-year follow-up study of a cross-section of the adult danish
dividuals seek medical attention, making the management of chronic population. Pain 2004;108:154–62.
pain a major issue in clinical practice. Left untreated, chronic pain can [18] Elliott AM, Smith BH, Penny KI, Smith WC, Chambers WA. The epidemiology of
lead to diminished quality of life and socioeconomic difficulties. chronic pain in the community. Lancet 1999;354:1248–52.
[19] Kuehn BM. Opioid prescriptions soar: increase in legitimate use as well as abuse.
Opioids continue to be the mainstay of chronic pain management. JAMA 2007;297:249–51.
However, opioid use can lead to abuse, diversion and physical depen- [20] National Center for Health Statistics. Health, United States, 2012: with Special Fea-
dence, meaning that opioids may not be suitable for all patients. An indi- ture on Emergency Care. Selected prescription drug classes used in the past 30
days, by sex and age: United States, selected years 1988–1994 through 2007–
vidual’s risk for abuse should be considered, by way of a detailed history, 2010, Table 92; 2012. http://www.cdc.gov/nchs/hus/contents2012.htm#092.
before opioids are prescribed. Several non-opioid based therapies are [21] Stamer UM, Stuber F. Genetic factors in pain and its treatment. Curr Opin
now potentially available for these patients, such as triptans, ziconotides, Anaesthesiol 2007;20:478–84.
[22] Stamer UM, Zhang L, Stuber F. Personalized therapy in pain management: where
as well as novel therapies, such as treatment with cannabinoids and, in do we stand? Pharmacogenomics 2010;11:843–64.
the not-so-distant future, gene therapy or epigenetic-based approaches. [23] Klepstad P, Rakvag TT, Kaasa S, Holthe M, Dale O, Borchgrevink PC, et al. The 118
Adjuvant therapies with antidepressants, benzodiazepines or anticon- A N G polymorphism in the human mu-opioid receptor gene may increase
morphine requirements in patients with pain caused by malignant disease. Acta
vulsants can also be useful in managing pain, particularly in the treat-
Anaesthesiol Scand 2004;48:1232–9.
ment of anxiety and depression, which are commonly associated with [24] Lalovic B, Kharasch E, Hoffer C, Risler L, Liu-Chen LY, Shen DD. Pharmacokinetics
chronic pain. and pharmacodynamics of oral oxycodone in healthy human subjects: role of
Genetic variability can have profound effects on drug metabolism circulating active metabolites. Clin Pharmacol Ther 2006;79:461–79.
[25] Kirchheiner J, Schmidt H, Tzvetkov M, Keulen JT, Lotsch J, Roots I, et al. Pharmaco-
and contribute to the inter-individual diversity in responses to pain kinetics of codeine and its metabolite morphine in ultra-rapid metabolizers due to
medications. However, there is a paucity of evidence for the benefits cyp2d6 duplication. Pharmacogenomics J 2007;7:257–65.
of pharmacogenetic testing in the context of pain management. Drug [26] Světlík S, Hronova K, Bakhouche H, Matouskova O, Slanar O. Pharmacogenetics of
chronic pain and its treatment. Mediators Inflamm 2013;2013:864319.
metabolism and responses are affected by many factors, including [27] Rendic S, Di Carlo FJ. Human cytochrome p450 enzymes: a status report summarizing
pharmacogenetics, with genetics offering only a partial explanation of their reactions, substrates, inducers, and inhibitors. Drug Metab Rev 1997;29:413–580.
an individual’s response. [28] Eichelbaum M, Ingelman-Sundberg M, Evans WE. Pharmacogenomics and individ-
ualized drug therapy. Annu Rev Med 2006;57:119–37.
At this point in time, therapeutic drug monitoring, through plasma [29] Wang B, Yang LP, Zhang XZ, Huang SQ, Bartlam M, Zhou SF. New insights into the
drug measurements, may be more useful than pharmacogenetics in structural characteristics and functional relevance of the human cytochrome P450
preventing adverse drug reactions to pain medications, while ensuring 2D6 enzyme. Drug Metab Rev 2009;41:573–643.
[30] Zhou SF, Liu JP, Chowbay B. Polymorphism of human cytochrome p450 enzymes
effective analgesia. Definitive, mass spectrometry based methods, and its clinical impact. Drug Metab Rev 2009;41:89–295.
capable of measuring parent drug and metabolite levels, are the most [31] Johansson I, Lundqvist E, Bertilsson L, Dahl ML, Sjoqvist F, Ingelman-Sundberg M.
useful assays for this purpose. Drug half-life can be a functional marker Inherited amplification of an active gene in the cytochrome p450 cyp2d locus as
a cause of ultrarapid metabolism of debrisoquine. Proc Natl Acad Sci U S A
of the cumulative effect of drug–drug interactions as well as pathophys-
1993;90:11825–9.
iological interactions that may affect the treatment drug’s disposition. [32] Yeo RK, Rostami-Hodjegan A, Tucker GT. Abundance of cytochromes P450 in
Currently, monitoring of pain management patients, if performed at human liver: a meta-analysis. Br J Clin Pharmacol 2004;57(5):687–8.
all, is largely through urine drug measurements, which do not correlate [33] Gasche Y, Daali Y, Fathi M, Chiappe A, Cottini S, Dayer P, et al. Codeine intoxication
associated with ultrarapid CYP2D6 metabolism. N Engl J Med 2004;351:2827–31.
with serum drug concentrations therefore limiting their use in monitor- [34] Koren G, Cairns J, Chitayat D, Gaedigk A, Leeder SJ. Pharmacogenetics of morphine
ing efficacy and toxicity. Direct determination of serum or plasma drug poisoning in a breastfed neonate of a codeine-prescribed mother. Lancet
levels may provide a more clinically useful, if logistically more difficult, 2006;368:704.
[35] Health Canada. Important safety information on tylenol with codeine in nursing
avenue for optimization of drug therapy, particularly in the context of mothers and ultra-rapid metabolizers of codeine - for health professionals; 2008.
chronic pain management. [36] FDA warning on codeine use by nursing mothers. http://www.fda.gov/NewsEvents/
Newsroom/PressAnnouncements/2007/ucm108968.htm; 2007 [Accessed 21 January
2014].
References [37] Pilotto A, Franceschi M, Vitale DF, Zaninelli A, Masotti G, Rengo F, et al. Upper gas-
trointestinal symptoms and therapies in elderly out-patients, users of non-selective
[1] Bonica JJ. The need of a taxonomy. Pain 1979;6:247–8. nsaids or coxibs. Aliment Pharmacol Ther 2005;22:147–55.
[2] Furlan AD, Reardon R, Weppler C, Group NOUG. Opioids for chronic noncancer [38] Visser LE, van Schaik RH, van VM, Trienekens PH, De Smet PA, Vulto AG, et al. Allelic
pain: a new Canadian practice guideline. Can Med Assoc J 2010;182:923–30. variants of cytochrome P450 2C9 modify the interaction between nonsteroidal

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx 17

anti-inflammatory drugs and coumarin anticoagulants. Clin Pharmacol Ther [68] Grosser T, Fries S, FitzGerald GA. Biological basis for the cardiovascular conse-
2005;77:479–85. quences of cox-2 inhibition: therapeutic challenges and opportunities. J Clin Invest
[39] Wadelius M, Chen LY, Lindh JD, Eriksson N, Ghori MJ, Bumpstead S, et al. The 2006;116:4–15.
largest prospective warfarin-treated cohort supports genetic forecasting.[erratum [69] Mukherjee D. Does a coxib-associated thrombotic risk limit the clinical use of the
appears in blood. 2014 feb 13;123(7):1113]. Blood 2009;113:784–92. compounds as analgesic anti-inflammatory drugs? Arguments in favor. Thromb
[40] Kirchheiner J, Stormer E, Meisel C, Steinbach N, Roots I, Brockmoller J. Influence of Haemost 2006;96:407–12.
CYP2C9 genetic polymorphisms on pharmacokinetics of celecoxib and its metabo- [70] Antman EM, Bennett JS, Daugherty A, Furberg C, Roberts H, Taubert KA, et al. Use of
lites. Pharmacogenetics 2003;13:473–80. nonsteroidal antiinflammatory drugs: an update for clinicians: a scientific state-
[41] Pilotto A, Seripa D, Franceschi M, Scarcelli C, Colaizzo D, Grandone E, et al. Genetic ment from the American Heart Association. Circulation 2007;115:1634–42.
susceptibility to nonsteroidal anti-inflammatory drug-related gastroduodenal [71] Jones VM. Acetaminophen injection: a review of clinical information. J Pain Palliat
bleeding: role of cytochrome P450 2C9 polymorphisms. Gastroenterology Care Pharmacother 2011;25:340–9.
2007;133:465–71. [72] Portenoy RK. Treatment of cancer pain. Lancet 2011;377:2236–47.
[42] Zwisler ST, Enggaard TP, Noehr-Jensen L, Mikkelsen S, Verstuyft C, Becquemont L, [73] Sawe J, Svensson JO, Rane A. Morphine metabolism in cancer patients on increasing
et al. The antinociceptive effect and adverse drug reactions of oxycodone in oral doses–no evidence for autoinduction or dose-dependence. Br J Clin Pharmacol
human experimental pain in relation to genetic variations in the oprm1 and 1983;16:85–93.
abcb1 genes. Fundam Clin Pharmacol 2010;24:517–24. [74] Koch T, Hollt V. Role of receptor internalization in opioid tolerance and depen-
[43] Campa D, Gioia A, Tomei A, Poli P, Barale R. Association of abcb1/mdr1 and dence. Pharmacol Ther 2008;117:199–206.
oprm1 gene polymorphisms with morphine pain relief. Clin Pharmacol Ther [75] Ferguson SS. Evolving concepts in g protein-coupled receptor endocytosis: the role
2008;83:559–66. in receptor desensitization and signaling. Pharmacol Rev 2001;53:1–24.
[44] Levran O, O'Hara K, Peles E, Li D, Barral S, Ray B, et al. ABCB1 (MDR1) genetic [76] von Zastrow, Svingos A, Haberstock-Debic H, Evans C. Regulated endocytosis of
variants are associated with methadone doses required for effective treatment of opioid receptors: cellular mechanisms and proposed roles in physiological adapta-
heroin dependence. Hum Mol Genet 2008;17:2219–27. tion to opiate drugs. Curr Opin Neurobiol 2003;13:348–53.
[45] Park HJ, Shinn HK, Ryu SH, Lee HS, Park CS, Kang JH. Genetic polymorphisms in [77] Bailey CP, Connor M. Opioids: cellular mechanisms of tolerance and physical de-
the ABCB1 gene and the effects of fentanyl in Koreans. Clin Pharmacol Ther pendence. Curr Opin Pharmacol 2005;5:60–8.
2007;81:539–46. [78] Mercadante S. Prospects and challenges in opioid analgesia for pain management.
[46] Lee YS, Kim H, Wu TX, Wang XM, Dionne RA. Genetically mediated interindividual Curr Med Res Opin 2011;27:1741–3.
variation in analgesic responses to cyclooxygenase inhibitory drugs. Clin [79] Manchikanti L, Ailinani H, Koyyalagunta D, Datta S, Singh V, Eriator I, et al. A sys-
Pharmacol Ther 2006;79:407–18. tematic review of randomized trials of long-term opioid management for chronic
[47] Janicki PK, Schuler G, Francis D, Bohr A, Gordin V, Jarzembowski T, et al. A genetic non-cancer pain. Pain Physician 2011;14:91–121.
association study of the functional A118G polymorphism of the human mu-opioid [80] Al-Sayed AA, Al-Numay AM. Update and review on the basics of pain management.
receptor gene in patients with acute and chronic pain. Anesth Analg Neurosciences 2011;16:203–12.
2006;103:1011–7. [81] Martin WR. Pharmacology of opioids. Pharmacol Rev 1983;35:283–323.
[48] Fillingim RB, Kaplan L, Staud R, Ness TJ, Glover TL, Campbell CM, et al. The a118g [82] Bohnert AS, Valenstein M, Bair MJ, Ganoczy D, McCarthy JF, Ilgen MA, et al. Associ-
single nucleotide polymorphism of the mu-opioid receptor gene (OPRM1) is ation between opioid prescribing patterns and opioid overdose-related deaths.
associated with pressure pain sensitivity in humans. J Pain 2005;6:159–67. JAMA 2011;305:1315–21.
[49] Landau R, Kern C, Columb MO, Smiley RM, Blouin JL. Genetic variability of the mu- [83] Chang G, Chen L, Mao J. Opioid tolerance and hyperalgesia. Med Clin North Am
opioid receptor influences intrathecal fentanyl analgesia requirements in laboring 2007;91:199–211.
women. Pain 2008;139:5–14. [84] Jamison RN, Serraillier J, Michna E. Assessment and treatment of abuse risk in opi-
[50] Rakvag TT, Klepstad P, Baar C, Kvam TM, Dale O, Kaasa S, et al. The val158met poly- oid prescribing for chronic pain. Pain Res Treat 2011;2011:941808.
morphism of the human catechol-O-methyltransferase (COMT) gene may influ- [85] Savage SR. Management of opioid medications in patients with chronic pain and
ence morphine requirements in cancer pain patients. Pain 2005;116:73–8. risk of substance misuse. Curr Psychiatry Rep 2009;11:377–84.
[51] Reyes-Gibby CC, Shete S, Rakvag T, Bhat SV, Skorpen F, Bruera E, et al. Exploring [86] Bair MJ, Robinson RL, Katon W, Kroenke K. Depression and pain comorbidity: a lit-
joint effects of genes and the clinical efficacy of morphine for cancer pain: erature review. Arch Intern Med 2003;163:2433–45.
OPRM1 and COMT gene. Pain 2007;130:25–30. [87] Cupp M. New oxycodone formulation: Oxyneo. Pharmaacist's Letter/Prescriber's
[52] Chen JF, Aloyo VJ, Weiss B. Continuous treatment with the d2 dopamine receptor Letter; 2012.
agonist quinpirole decreases D2 dopamine receptors, D2 dopamine receptor mes- [88] Houde RW. Analgesic effectiveness of the narcotic agonist-antagonists. Br J Clin
senger RNA and proenkephalin messenger RNA, and increases mu opioid receptors Pharmacol 1979;7:297S–308S.
in mouse striatum. Neuroscience 1993;54:669–80. [89] Vallejo R, Barkin RL, Wang VC. Pharmacology of opioids in the treatment of chronic
[53] Steiner H, Gerfen CR. Role of dynorphin and enkephalin in the regulation of striatal pain syndromes. Pain Physician 2011;14:E343–60.
output pathways and behavior. Exp Brain Res 1998;123:60–76. [90] Hutchinson MR, Menelaou A, Foster DJ, Coller JK, Somogyi AA. Cyp2d6 and cyp3a4
[54] Ross JR, Riley J, Taegetmeyer AB, Sato H, Gretton S, du Bois RM, et al. Genetic vari- involvement in the primary oxidative metabolism of hydrocodone by human liver
ation and response to morphine in cancer patients: catechol-O-methyltransferase microsomes. Br J Clin Pharmacol 2004;57:287–97.
and multidrug resistance-1 gene polymorphisms are associated with central side [91] Sansone RA, Sansone LA. Tramadol: seizures, serotonin syndrome, and
effects. Cancer 2008;112:1390–403. coadministered antidepressants. Psychiatry (Edgmont) 2009;6:17–21.
[55] Kadiev E, Patel V, Rad P, Thankachan L, Tram A, Weinlein M, et al. Role of pharma- [92] Stahl SM. Mechanism of action of serotonin selective reuptake inhibitors. Serotonin
cogenetics in variable response to drugs: focus on opioids. Expert Opin Drug Metab receptors and pathways mediate therapeutic effects and side effects. J Affect Disord
Toxicol 2008;4:77–91. 1998;51:215–35.
[56] Fishbain DA, Fishbain D, Lewis J, Cutler RB, Cole B, Rosomoff HL, et al. Genetic test- [93] Nelson EM, Philbrick AM. Avoiding serotonin syndrome: the nature of the interac-
ing for enzymes of drug metabolism: does it have clinical utility for pain medicine tion between tramadol and selective serotonin reuptake inhibitors. Ann
at the present time? A structured review. Pain Med 2004;5:81–93. Pharmacother 2012;46:1712–6.
[57] Flockhart DA. Drug interactions: cytochrome p450 drug interaction. http://medi- [94] Dayer P, Desmeules J, Collart L. Pharmacologie du tramadol. Drugs 1997;53(Suppl.-
cine.iupui.edu/clinpharm/ddis/clinical-table/; 2014 . [Accessed 28 January 2014]. 24).
[58] Ablin JN, Buskila D. Personalized treatment of pain. Curr Rheumatol Rep [95] Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clin Pharmacokinet
2013;15:298. 2004;43:879–923.
[59] Cherny N, Ripamonti C, Pereira J, Davis C, Fallon M, McQuay H, et al. Strategies to [96] Sawe J, Dahlstrom B, Paalzow L, Rane A. Morphine kinetics in cancer patients. Ann
manage the adverse effects of oral morphine: an evidence-based report. J Clin Pharmacother 1981;30:629–35.
Oncol 2001;19:2542–54. [97] Cregg R, Russo G, Gubbay A, Branford R, Sato H. Pharmacogenetics of analgesic
[60] Rane A, Sawe J, Dahlstrom B, Paalzow L, Kager L. Pharmacological treatment of can- drugs. Br Pain 2013;7:189–206.
cer pain with special reference to the oral use of morphine. Acta Anaesthesiol [98] Berkenstadt H, Segal E, Mayan H, Almog S, Rotenberg M, Perel A, et al. The pharma-
Scand 1982:74–103 [Supplementum]. cokinetics of morphine and lidocaine in critically ill patients. Intensive Care Med
[61] World Health Organization Cancer pain relief. http://www.whocancerpain.wisc. 1999;25:110–2.
edu/?q=node/169; 1986 . [Accessed 21 January 2014]. [99] Smith MT. Neuroexcitatory effects of morphine and hydromorphone: evidence impli-
[62] Vargas-Schaffer G. Is the who analgesic ladder still valid? Twenty-four years of cating the 3-glucuronide metabolites. Clin Exp Pharmacol Physiol 2000;27:524–8.
experience. Can Fam Physician 2010;56:514–7. [100] Mather LE. Clinical pharmacokinetics of fentanyl and its newer derivatives. Clin
[63] Varrassi G. Severe chronic pain–the reality of treatment in europe. Curr Med Res Pharmacokinet 1983;8:422–46.
Opin 2011;27:2063–4. [101] Reynolds L, Rauck R, Webster L, DuPen S, Heinze E, Portenoy R, et al. Relative
[64] Rollins DE, von BC, Glaumann H, Moldeus P, Rane A. Acetaminophen: potentially analgesic potency of fentanyl and sufentanil during intermediate-term infusions in
toxic metabolite formed by human fetal and adult liver microsomes and isolated patients after long-term opioid treatment for chronic pain. Pain 2004;110:182–8.
fetal liver cells. Science 1979;205:1414–6. [102] Kosek E, Jensen KB, Lonsdorf TB, Schalling M, Ingvar M. Genetic variation
[65] Mitchell JR, Jollow DJ, Potter WZ, Davis DC, Gillette JR, Brodie BB. Acetaminophen- in the serotonin transporter gene (5-HTTLPR, RS25531) influences the analge-
induced hepatic necrosis. I. Role of drug metabolism. J Pharmacol Exp Ther sic response to the short acting opioid remifentanil in humans. Mol Pain
1973;187:185–94. 2009;5:37.
[66] You K. Salicylate and mitochondrial injury in Reye's syndrome. Science [103] Burkle H, Dunbar S, Van AH. Remifentanil: a novel, short-acting, mu-opioid. Anesth
1983;221:163–5. Analg 1996;83:646–51.
[67] Reye RDK, Morgan G, Baral J. Encephalopathy and fatty degeneration of the viscera [104] Anderson D. A review of systemic opioids commonly used for labor pain relief. J
a disease entity on childhood. Lancet 1963;2(7311):749–52. Midwifery Womens Health 2011;56:222–39.

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
18 B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx

[105] Leong WL, Sng BL, Sia AT. A comparison between remifentanil and meperidine for [142] Ware M, Beaulieu P. Cannabinoids for the treatment of pain: an update on recent
labor analgesia: A systematic review. Anesth Analg 2011;113:818–25. clinical trials. Pain Res Manag 2005;10:27A–30A.
[106] Kan RE, Hughes SC, Rosen MA, Kessin C, Preston PG, Lobo EP. Intravenous [143] de Vries M, van Rijckevorsel DC, Wilser-Smith OH, van Goor H. Dronabinol
remifentanil: placental transfer, maternal and neonatal effects. Anesthesiology and chronic pain: importance of mechanistic considerations. Expert Opin
1998;88:1467–74. Pharmacother 2014;12:1–10.
[107] Fredheim OM, Borchgrevink PC, Klepstad P, Kaasa S, Dale O. Long term methadone for [144] Clark AJ, Lynch ME, Ware M, Beaulieu P, McGilveray IJ, Gourlay D. Guidelines for
chronic pain: a pilot study of pharmacokinetic aspects. Eur J Pain 2007;11:599–604. the use of cannabinoid compounds in chronic pain. Pain Res Manag
[108] Lugo RA, Satterfield KL, Kern SE. Pharmacokinetics of methadone. J Pain Palliat Care 2005;10:44A–6A.
Pharmacother 2005;19:13–24. [145] Donoghue K, Doody GA, Murray RM, Jones PB, Morgan C, Dazzan P, et al. Cannabis
[109] Eap CB, Buclin T, Baumann P. Interindividual variability of the clinical pharmacoki- use, gender and age of onset of schizophrenia: data from the ÆSOP Study. Psychi-
netics of methadone: implications for the treatment of opioid dependence. Clin atry Res 2014;215:528–32.
Pharmacokinet 2002;41:1153–93. [146] Olivera BM. Conus peptides: biodiversity-based discovery and exogenomics. J Biol
[110] Kapur BM, Hutson JR, Chibber T, Luk A, Selby P. Methadone: a review of drug–drug Chem 2006;281:31173–7.
and pathophysiological interactions. Crit Rev Clin Lab Sci 2011;48:171–95. [147] Schmidtko A, Lotsch J, Freynhagen R, Geisslinger G. Ziconotide for treatment of
[111] Perrin-Terrin A, Pathak A, Lapeyre-Mestre M. Qt interval prolongation: prevalence, severe chronic pain. Lancet 2010;375:1569–77.
risk factors and pharmacovigilance data among methadone-treated patients in [148] Wermeling D, Drass M, Ellis D, Mayo M, McGuire D, O'Connell D, et al. Pharmaco-
france. Fundam Clin Pharmacol 2011;25:503–10. kinetics and pharmacodynamics of intrathecal ziconotide in chronic pain patients.
[112] Johnson RE, Fudala PJ, Payne R. Buprenorphine: considerations for pain manage- J Clin Pharmacol 2003;43:624–36.
ment. J Pain Symptom Manage 2005;29:297–326. [149] Geranton SM. Targeting epigenetic mechanisms for pain relief. Curr Opin
[113] Pergolizzi J, Aloisi AM, Dahan A, Filitz J, Langford R, Likar R, et al. Current knowledge of Pharmacol 2012;12:35–41.
buprenorphine and its unique pharmacological profile. Pain Pract 2010;10:428–50. [150] Tajerian M, Alvarado S, Millecamps M, Dashwood T, Anderson KM, Haglund L, et al.
[114] Leow KP, Cramond T, Smith MT. Pharmacokinetics and pharmacodynamics of oxy- DNA methylation of sparc and chronic low back pain. Mol Pain 2011;7:65.
codone when given intravenously and rectally to adult patients with cancer pain. [151] Uchida H, Sasaki K, Ma L, Ueda H. Neuron-restrictive silencer factor causes epige-
Anesth Analg 1995;80:296–302. netic silencing of Kv4.3 gene after peripheral nerve injury. Neuroscience
[115] Sloan P. Review of oral oxymorphone in the management of pain. Ther Clin Risk 2010;166:1–4.
Manage 2008;4:777–87. [152] Uchida H, Ma L, Ueda H. Epigenetic gene silencing underlies c-fiber dysfunctions in
[116] Batterman RC. Clinical effectiveness and safety of a new synthetic analgesic drug, neuropathic pain. J Neurosci 2010;30:4806–14.
demerol. Arch Intern Med 1943;71:345–56. [153] Robak T. New nucleoside analogs for patients with hematological malignancies.
[117] Latta KS, Ginsberg B, Barkin RL. Meperidine: a critical review. Am J Ther Expert Opin Investig Drugs 2011;20:343–59.
2002;9:53–68. [154] Bai G, Wei D, Zou S, Ren K, Dubner R. Inhibition of class ii histone deacetylases in
[118] Saravanakumar K, Garstang JS, Hasan K. Intravenous patient-controlled analgesia the spinal cord attenuates inflammatory hyperalgesia. Mol Pain 2010;6:51.
for labour: a survey of UK practice. Int J Obstet Anesth 2007;16:221–5. [155] Agrawal RP, Goswami J, Jain S, Kochar DK. Management of diabetic neuropathy by
[119] Tveit TO, Halvorsen A, Rosland JH. Analgesia for labour: a survey of Norwegian sodium valproate and glyceryl trinitrate spray: a prospective double-blind
practice — with a focus on parenteral opioids. Acta Anaesthesiol Scand randomized placebo-controlled study. Diabetes Res Clin Pract 2009;83:371–8.
2009;53:794–9. [156] Handy CR, Krudy C, Boulis N. Gene therapy: a potential approach for cancer pain.
[120] Prommer E. Levorphanol: the forgotten opioid. Support Care Cancer 2007;15:259–64. Pain Res Treat 2011:987597.
[121] Sansone RA, Sansone LA. Pain, pain, go away: antidepressants and pain manage- [157] Fink DJ, Wechuck J, Mata M, Glorioso JC, Goss J, Krisky D, et al. Gene therapy for
ment. Psychiatry (Edgmont) 2008;5:16–9. pain: results of a phase i clinical trial. Ann Neurol 2011;70:207–12.
[122] Gureje O, Von KM, Simon GE, Gater R. Persistent pain and well-being: a World [158] Manchikanti L, Damron KS, McManus CD, Barnhill RC. Patterns of illicit drug use
Health Organization study in primary care. JAMA 1998;280:147–51. and opioid abuse in patients with chronic pain at initial evaluation: a prospective,
[123] Eardley W, Toth C. An open-label, non-randomized comparison of venlafaxine and observational study. Pain Physician 2004;7:431–7.
gabapentin as monotherapy or adjuvant therapy in the management of neuropath- [159] Manchikanti L, Manchukonda R, Pampati V, Damron KS, Brandon DE, Cash KA, et al.
ic pain in patients with peripheral neuropathy. J Pain Res 2010;1:33–49. Does random urine drug testing reduce illicit drug use in chronic pain patients
[124] Moulin DE, Clark AJ, Gilron I, Ware MA, Watson CP, Sessle BJ, et al. Pharmacological receiving opioids? Pain Physician 2006;9:123–9.
management of chronic neuropathic pain - consensus statement and guidelines [160] Manchikanti L, Atluri S, Trescot AM, Giordano J. Monitoring opioid adherence in chron-
from the canadian pain society. Pain Res Manag 2007;12:13–21. ic pain patients: tools, techniques, and utility. Pain Physician 2008;11(Suppl.-80).
[125] Jensen TS. Anticonvulsants in neuropathic pain: rationale and clinical evidence. Eur [161] Nafziger AN, Bertino Jr JS. Utility and application of urine drug testing in chronic
J Pain 2002;6:A61–8. pain management with opioids. Clin J Pain 2009;25:73–9.
[126] Bagheri SC, Farhidvash F, Perciaccante VJ. Diagnosis and treatment of patients with [162] Kapur BM, Verjee Z, Anderson M, Geisbrecht E. Urinary eddp (methadone metabolite)
trigeminal neuralgia. J Am Dent Assoc 2004;135:1713–37. as a marker of compliance and diversion of methadone. Ther Drug Monit 1995;17:388.
[127] Dolder CR, Nealy KL. The efficacy and safety of newer anticonvulsants in patients [163] Jannetto PJ, Bratanow NC. Pain management in the 21st century: utilization of
with dementia. Drugs Aging 2012;29:627–37. pharmacogenomics and therapeutic drug monitoring. Expert Opin Drug Metab
[128] Rose MA, Kam PC. Gabapentin: pharmacology and its use in pain management. Toxicol 2011;7:745–52.
Anaesthesia 2002;57:451–62. [164] Levy G, Tsuchiya T. Salicylate accumulation kinetics in man. N Engl J Med
[129] Ward KE, Archambault R, Mersfelder TL. Severe adverse skin reactions to nonste- 1972;287:430–2.
roidal antiinflammatory drugs: a review of the literature. Am J Health Syst Pharm [165] Adams SS, McCullough KF, Nicholson JS. The pharmacological properties of ibupro-
2010;67:206–13. fen, an anti-inflammatory, analgesic and antipyretic agent. Arch Int Pharmacodyn
[130] Gentile G, Borro M, Simmaco M, Missori S, Lala N, Martelletti P. Gene polymor- Ther 1969;178:115–29.
phisms involved in triptans pharmacokinetics and pharmacodynamics in migraine [166] Alvan G, Orme M, Bertilsson L, Ekstrand R, Palmer L. Pharmacokinetics of indo-
therapy. Expert Opin Drug Metab Toxicol 2011;7:39–47. methacin. Clin Pharmacol Ther 1975;18:364–73.
[131] Loder E. Triptan therapy in migraine. N Engl J Med 2010;363:63–70. [167] Morris ME, Levy G. Renal clearance and serum protein binding of acetaminophen
[132] Gawel MJ, Worthington I, Maggisano A. A systematic review of the use of triptans and its major conjugates in humans. J Pharm Sci 1984;73:1038–41.
in acute migraine. Can J Neurol Sci 2001;28:30–41. [168] Davies NM, McLachlan AJ, Day RO, Williams KM. Clinical pharmacokinetics and
[133] Dellemijn PL, Fields HL. Do benzodiazepines have a role in chronic pain manage- pharmacodynamics of celecoxib: a selective cyclo-oxygenase-2 inhibitor. Clin
ment? Pain 1994;57:137–52. Pharmacokinet 2000;38:225–42.
[134] Manzanares J, Julian M, Carrascosa A. Role of the cannabinoid system in pain con- [169] Baselt R. Disposition of toxic drugs and chemicals in man. Ninth Edition ed. Seal
trol and therapeutic implications for the management of acute and chronic pain Beach, California: Biomedical Publications; 2011.
episodes. Curr Neuropharmacol 2006;4:239–57. [170] Blaine JD. Buprenorphine: an alternative treatment for opioid dependence. In: Jack
[135] Walker JM, Huang SM. Cannabinoid analgesia. Pharmacol Ther 2002;95:127–35. D, Blaine P, editors. 5600 Fishers Lane, Rockville, MD 20857, USA: National Institute
[136] Leung L. Cannabis and its derivatives: review of medical use. J Am Board Fam Med on Drug Abuse; 1992.
2011;24:452–62. [171] Bullingham RE, McQuay HJ, Moore A, Bennett MR. Buprenorphine kinetics. Clin
[137] Abrams DI, Jay CA, Shade SB, Vizoso H, Reda H, Press S, et al. Cannabis in painful Pharmacol Ther 1980;28:667–72.
hiv-associated sensory neuropathy: a randomized placebo-controlled trial. Neurol- [172] Han T, Harmatz JS, Greenblatt DJ, Martyn JA. Fentanyl clearance and volume of
ogy 2007;68:515–21. distribution are increased in patients with major burns. J Clin Pharmacol
[138] Ellis RJ, Toperoff W, Vaida F, van den BG, Gonzales J, Gouaux B, et al. Smoked me- 2007;47:674–80.
dicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. [173] Parab PV, Ritschel WA, Coyle DE, Gregg RV, Denson DD. Pharmacokinetics of
Neuropsychopharmacology 2009;34:672–80. hydromorphone after intravenous, peroral and rectal administration to human
[139] Rog DJ, Nurmikko TJ, Friede T, Young CA. Randomized, controlled trial of cannabis- subjects. Biopharm Drug Dispos 1988;9:187–99.
based medicine in central pain in multiple sclerosis. Neurology 2005;65:812–9. [174] Ramirez J, Innocenti F, Schuetz EG, Flockhart DA, Relling MV, Santucci R, et al.
[140] Vaney C, Heinzel-Gutenbrunner M, Jobin P, Tschopp F, Gattlen B, Hagen U, et al. CYP2B6, CYP3A4, and CYP2C19 are responsible for the in vitro n-demethylation
Efficacy, safety and tolerability of an orally administered cannabis extract in the of meperidine in human liver microsomes. Drug Metab Dispos 2004;32:930–6.
treatment of spasticity in patients with multiple sclerosis: a randomized, double- [175] Pond SM, Tong T, Benowitz NL, Jacob P, Rigod J. Presystemic metabolism of meper-
blind, placebo-controlled, crossover study. Mult Scler 2004;10:417–24. idine to normeperidine in normal and cirrhotic subjects. Clin Pharmacol Ther
[141] Zajicek JP, Sanders HP, Wright DE, Vickery PJ, Ingram WM, Reilly SM, et al. Canna- 1981;30:183–8.
binoids in multiple sclerosis (cams) study: safety and efficacy data for 12 months [176] Schulz P, Turner-Tamiyasu K, Smith G, Giacomini KM, Blaschke TF. Amitriptyline
follow up. J Neurol Neurosurg Psychiatry 2005;76:1664–9. disposition in young and elderly normal men. Clin Pharmacol Ther 1983;33:360–6.

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
B.M. Kapur et al. / Clinical Biochemistry xxx (2014) xxx–xxx 19

[177] Stewart JJ, Berkel HJ, Parish RC, Simar MR, Syed A, Bocchini Jr JA, et al. Single-dose [183] Mesdjian E, Seree E, Charvet B, Mirrione A, Bourgarel-Rey V, Desobry A, et al.
pharmacokinetics of bupropion in adolescents: effects of smoking status and Metabolism of carbamazepine by CYP3A6: a model for in vitro drug interactions
gender. J Clin Pharmacol 2001;41:770–8. studies. Life Sci 1999;64:827–35.
[178] Rampono J, Kristensen JH, Hackett LP, Paech M, Kohan R, Ilett KF. Citalopram and [184] Rawlins MD, Collste P, Bertilsson L, Palmer L. Distribution and elimination kinetics
demethylcitalopram in human milk; distribution, excretion and effects in breast of carbamazepine in man. Eur J Clin Pharmacol 1975;8:91–6.
fed infants. Br J Clin Pharmacol 2000;50:263–8. [185] McLean MJ. Clinical pharmacokinetics of gabapentin. Neurology 1994;44(Suppl.-22).
[179] Rudorfer MV, Lane EA, Chang WH, Zhang MD, Potter WZ. Desipramine pharmaco- [186] Chan V, Morris RG, Ilett KF, Tett SE. Population pharmacokinetics of lamotrigine.
kinetics in chinese and caucasian volunteers. Br J Clin Pharmacol 1984;17:433–40. Ther Drug Monit 2001;23:630–5.
[180] Deepak P, Kumar TN. Duloxetine - pharmacological aspects. Int J Biol Med Res [187] Bergen DC. Pharmacokinetics of phenytoin: reminders and discoveries. Epilepsy
2011;2:589–92. Curr 2009;9:102–4.
[181] Altamura AC, Moro AR, Percudani M. Clinical pharmacokinetics of fluoxetine. Clin [188] Cuttle L, Munns AJ, Hogg NA, Scott JR, Hooper WD, Dickinson RG, et al. Phenytoin
Pharmacokinet 1994;26:201–14. metabolism by human cytochrome P450: involvement of P450 3A and 2C forms
[182] Ellingrod VL, Perry PJ. Venlafaxine: a heterocyclic antidepressant. Am J Hosp Pharm in secondary metabolism and drug-protein adduct formation. Drug Metab Dispos
1994;51:3033–46. 2000;28:945–50.

Please cite this article as: Kapur BM, et al, Pharmacogenetics of chronic pain management, Clin Biochem (2014), http://dx.doi.org/10.1016/
j.clinbiochem.2014.05.065
View publication stats

You might also like