AJA Asian Journal of Anesthesiology

Advancing, Capability, Improving lives

Review Article
Volume 54, Issue 1, Pages 24-30
Tai-Ming Ko 1.2 , Chih-Shung Wong 3 , Jer-Yuarn Wu 1.4 , Yuan-Tsong Chen 1.5
3600 Views


Abstract

Pharmacogenomics aims to unravel the way that human genetic variation affects drug efficacy and toxicity. Genome-wide association studies and candidate gene findings suggest that genetic approaches may help choose the most appropriate drug and dosage while preventing adverse drug reactions (ADRs). Pain is an unpleasant feeling that usually results from tissue damage. The management of different types of pain (acute, chronic, inflammatory, neuropathic, or cancer) is challenging. Currently, drug intervention is the first-line therapy for resolving pain. However, differences in drug efficacy between individuals are common with pain medications. Moreover, some patients experience ADRs after being treated with specific pain drugs. This review discusses the use of drugs for pain management in the context of the recent pharmacogenomic studies on ADRs and drug efficacy.

Keywords

adverse drug reactionsgenome wide association studiespersonalized medicinepharmacogenomics;


1. Introduction

A medication with proven efficacy and safety in a series of rigorous clinical trials could still fail to work in some patients or cause serious adverse drug reactions (ADRs).12 The interindividual responses to drugs are most likely affected by genetic variations, which can be divided into two types: (1) inherited variants (i.e., germ-line genetic variants); and (2) acquired variants (i.e., somatic mutation). Germ-line variants of genes encoding drug-metabolizing enzymes, drug transporters, drug targets, and human-leukocyte antigen (HLA) can affect individual response to medications. Somatic variants of genes are frequently associated with the development or progression of cancer, and affect the drug response of tumors that carry specific mutations, so called target therapy. Because of the impact of genetic variants on medication responses, how to give the “right drug” at the “right dose” for the “right patient” is a major goal in the era of precision medicine.34

2. Pharmacogenomics

Pharmacogenomics is the application of current technology for the precise determination of genetic variants that influence drug response, and to develop personalized strategies that maximize therapeutic efficacy and assure drug safety. Large-scale genome-wide association studies and smaller-scale studies with a candidate-gene approach, that are used to study genes involved in drug metabolism enzymes, drug transporters, drug receptors, and HLA, have helped advance our understanding of the underlying mechanisms of ADRs and drug efficacy (Figure 1). Next-generation whole-genome sequencing, which provides a diverse genome map of multiple populations, will be useful for the future of pharmacogenetic studies.5 These studies take into account ancestral genetic structure, complex haplotypes, gene–gene interactions, and rare variants. They aim to detect and replicate novel pharmacogenetic loci of clinical significance. Based on these studies, the United States Food and Drug Administrations (FDA) have relabeled over 100 approved drugs with genetic information. A list of valid genomic biomarkers for clinical guidance can be found on the FDA website “Table of Pharmacogenomic Biomarkers in Drug Labels” (http://www.fda.gov/Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics/ucm083378.htm). Some of these biomarkers have been implemented in medical practice.678

Figure 1.
Download full-size image
Figure 1. Pharmacogenetic testing for pain management. Pharmacogenetic tests provide information about a patient's likelihood to have an adverse drug reaction (ADR) and/or a therapeutic response to a medication before prescribing pain medication. For giving the right drugs and right doses for right patients, a precise therapeutic intervention (i.e., adjust drug dosage or avoid use the drug) should be based on the information of the pharmacogenetic tests.

3. Pain medication

Most clinical pain management options involve pharmacological interventions. Pain therapy has evolved over the years into a large specialty field. Ideal pain management approaches must provide adequate analgesia without excessive adverse effects. However, there are large interindividual differences in response to pain medications, concerning efficacy and the development of severe ADRs.910111213 Current pain management strategies are devised using the World Health Organization pain ladder, which begins with nonopioid medications, such as nonsteroidal anti-inflammatory drugs (NSAIDs), progressing to weak opioids, and culminating with strong opioids.9 Other pain medications include anticonvulsant drugs for neuralgia. Additionally, adjuvant therapies using antidepressant medications can aid in reducing chronic pain-associated anxiety.1415 In Taiwan, based on the National Health Insurance Research Database, we found the drugs that are most frequently used for pain management (Table 1Table 2).

The field of pain medications is a valuable one to study germ-line variants using a pharmacogenomic approach, because of the considerable repercussions of these medications on the biological, psychological, sociological, and economical welfare of patients. Genetic studies have identified several polymorphic loci that govern the pharmacodynamics and kinetics of analgesic drugs.161718 The aim of this review is to highlight recent advances in the pharmacogenomics of pain medicine.

3.1. Drug metabolism enzymes

The metabolism of drugs and other xenobiotics is often divided into multiple phases. Phase I enzymes are responsible for chemical modifications of the drugs. They include cytochrome P450 (CYP), cytochrome b5, and nicotinamide adenine dinucleotide phosphate-cytochrome P450 reductase. Phase II enzymes are involved in further conjugation of the active drug metabolites, some examples are glutathione S-transferases, aryl sulfatase, and uridine-glucuronosyltransferase. The hepatic CYPs are a multigene family of enzymes that play a critical role in the metabolism of many drugs, with each cytochrome isozyme displaying unique substrate specificities and susceptibility to induction and inhibition by exogenous chemicals. One of the most common CYPs involved in drug metabolism is cytochrome P450, family 2, subfamily D, polypeptide 6 (CYP2D6), whose metabolic rate can fluctuate by over 100-fold between the allelic variants expressed in different ethnic groups.19 For example, approximately 10% of the Caucasian population carries an autosomal recessive trait that yields a nonfunctional variant.20 Patients homozygous for this variant, known as poor metabolizers (PM), may have either a higher risk of adverse side effects because of drug overdose (i.e., tricyclic antidepressants or antiarrhythmics) or no drug efficacy due to poor transformation of the prodrug into its active metabolite. Conversely, in ultra-rapid metabolizers (UM), amplification of the CYP2D6 gene is correlated with enhanced enzyme activity and ultrarapid metabolism, which could decrease the optimal dosage for UM (Figure 1).212223

3.2. Opioid analgesics-codeine: CYP2D6

Opioids are the pillar of pharmacological pain management.24 They have been used in therapeutics for centuries, and there are > 30 opioids currently marketed worldwide. However, opioid over dosage is highly correlated with respiratory depression, a harmful and life-threatening adverse effect.25 Careful therapeutic drug monitoring of the plasma concentrations can help provide effective pain relief while minimizing adverse effects. Morphine and diamorphine have been shown to have a wider therapeutic range or “safety margin” than some other opioids. Since it is not possible to tell which patients will require low or high doses, commencement dose is always low, unless it is a therapy change from another strong opioid.26 Thus, opioid treatment requires an individualized approach to ensure safety. Ample studies have demonstrated that this individualized treatment might rely on pharmacogenomics. The future promise of pharmacogenetics involves a rational drug regimen that maximizes efficacy and minimizes adverse events. However, variation in responses to pain medication is partially explained or predicted by patient's genetics, and pharmacogenetic testing for opioids is not widely applied in current clinical practice.

Codeine is widely administered after operations and is used in certain drug combinations for acute and chronic pain management.2728 Because codeine is a less potent μ-opioid receptor agonist than morphine, it has been classified as a weak opioid.2930 Codeine is a prodrug with a low affinity for the opioid receptor and low intrinsic activity at the μ-opioid receptor. The initial metabolism of codeine is through glucuronidation with O-demethylation,32resulting in morphine formation. The O-demethylation of codeine to morphine is mediated by CYP2D6.33 UMs (CYP2D6*1/*1 and *1/*2) should avoid codeine due to its potential for toxicity (Table 3). The Clinical Pharmacogenetics Implementation Consortium guidelines have provided information for guiding the dosing of codeine based on CYP2D6 genotypes.34It is suggested to consider alternative analgesics for UMs such as morphine or a nonopioi

3.3. Opioid analgesics: drug receptors and transporters

In addition to the association between CYP2D6 (metabolism) and opioid response, the major candidate genetic contributors to opioid efficacy and adverse effects are ABCB1 (drug transporter) and OPRM1 (receptor interaction).232638394041 The μ-opioid receptor (OPRM1) is the primary binding site of action for many opioid drugs and for binding of beta-endorphins. The μ-opioid receptor is a transmembrane protein which belongs to the rhodopsin family of G-protein coupled receptors. The downstream signaling of μ-opioid receptors is through interactions with heterotrimeric G proteins.42 To date, over 100 genetic polymorphisms have been found in the OPRM1 gene. The most common studied nonsynonymous single nucleotide polymorphism is rs1799971, which is located in exon 1, and is a nucleotide change from adenosine (A) to guanosine (G) (A118G). Depending on the ethnicity, the polymorphism can be found in a proportion from 2% to nearly 50%. The μ-opioid receptor polymorphism was also found to be associated with opioid effects43. The functional study of OPRM1 revealed that the G allele (A118G) creates a novel CpG-methylation site. This methylation would suppress the increase of the OPRM1 expression level that takes place after a prolonged opioid administration. Patients with an inactive OPRMI allele might benefit from a k-agonist such as buprenorphine instead of a μ-agonist such as morphine.

3.4. Opioid analgesics-methadone: CYP3A4, CYP2B6, and CYP2D6

Methadone is an old opioid drug, having been discovered and developed during World War II.44 It is an opioid with a completely different chemical structure to morphine and oxycodone. Currently, methadone (including R- and S-methadone) are used as second-line opioids for the treatment of patients suffering from cancer pain.45 Methadone is a useful alternative in individuals who have had no responses to other opioids or suffering from adverse side effects (ADRs).464748 It has demonstrated its efficacy in neuropathic pain conditions,49 with several case reports highlighting this specific indication owing to methadone's N-methyl-D-aspartate-receptor agonistic properties.5051 Both methadone enantiomers (R- and S-methadone) bind to the noncompetitive site of the N-methyl-D-aspartate receptor. In addition, R-methadone proved to be a stronger μ-opioid receptor agonist than S-methadone. However, because the pharmacokinetics of methadone was still unclear, the methadone has a narrow therapeutic index.52Studies on the metabolism of methadone have mainly been carried out on cohorts of opioid addicts under methadone maintenance treatment. In vivoCYP3A4 and CYP2B6 are the major CYP isoforms involved in methadone metabolism, with CYP2D6 contributing to a minor extent, preferentially in metabolism of the R-enantiomer (Table 3).53

3.5. NSAIDs as nonopioid analgesics: CYP2C9

NSAIDs are a group of nonopioid analgesics commonly used for the treatment of acute pain after surgery or chronic pain. Although multimodal pain management by NSAIDS and opioids is important to enhance the efficacy of treatment and reduce the adverse effects, some adverse events including gastrointestinal side effects and cardiovascular side effects have been reported after long-term treatment.54 It implies that an interindividual variation limits the clinical utility and safety of NSAIDS. Based on the previous reports, CYP2C9 polymorphisms might play a significant role in the analgesic efficacy and toxicity of traditional NSAIDs (Table 3), such as flurbiprofen and celecoxib (Table 3).5455 For NSAIDs used as analgesics, treatment should start at half the lowest recommended dose in poor metabolizers (CYP2C9*3/*3 genotype) to avoid adverse cardiovascular and gastrointestinal events (Table 3). For traditional NSAIDs or selective cyclooxygenase-2 inhibitors, few data are currently available on the possible impact of these single nucleotide polymorphisms on analgesic efficacy.56

3.6. Cyclic antidepressant drugs as co-analgesics: CYP2D6 and CYP2C19

Cyclic antidepressants tackle depression by regulating the levels of neurotransmitters in the brain.5758 Currently, a major issue of the treatment with tricyclic antidepressants is their narrow therapeutic index, which results in their replacement by other antidepressants such as selective serotonin reuptake inhibitors.5960 However, because of their significant efficacy for pain management, cyclic antidepressants are frequently used as “co-analgesics” in the treatment of chronic pain, especially neuropathic pain61; they are also used for migraine prophylaxis. Some of the most commonly prescribed co-analgesics include amitriptyline, desipramine, imipramine, nortriptyline, doxepin, clomipramine, and protriptyline. Maprotiline (a tetracyclic compound) and amoxapine (a tricyclic dibenzoxazepine) are newer compounds that have a slightly different structure and toxicological profile.

Cyclic antidepressants are activated in the liver, after hydroxylation by CYP2D6 and demethylation by CYP2C19.62 CYP2D6 PMs have higher plasma concentrations of cyclic antidepressants than extensive metabolizers.63 PMs with CYP2D6*3/*3 genotypes should have the dose reduced by 60% to avoid arrhythmia and myelosuppression (Table 3). Therefore, the PMs have a higher risk of ADRs. By contrast, carriers of the CYP2D6 gene duplications would have ultrarapid metabolism of cyclic antidepressants, which may result in lower drug concentrations and poorer therapeutic responses.64 Thus, genetic variants may affect plasma concentrations of antidepressants, and dose adjustments before treatment are recommended.

4. Pharmacogenomics for pain medicine: immune-mediated drug hypersensitivity

4.1. Antiepileptic drugs for neuralgia: HLA association of drug-induced cutaneous ADRs

Antiepileptic drugs (AEDs) typically are used to control epilepsy or to treat some psychiatric disorders, e.g., bipolar disorder. These drugs, particularly carbamazepine, trileptal, lamotrigine, gabapentin, and topiramate, may also be used to treat some painful conditions, such as postherpetic neuralgia and fibromyalgia. However, anticonvulsants are a common cause of cutaneous ADRs (cADRs).65 The spectrum of cADRs ranges from a mild maculo–papular exanthema to life-threatening severe cutaneous reactions (SCARs) including drug rash with eosinophilia and systemic signs, Stevens–Johnson syndrome (SJS), and toxic epidermal necrolysis (TEN). More than 90% of SCARs occur within 2 months of AED use.66 The AEDs with risk of SCARs include carbamazepine, phenobarbital, phenytoin, and lamotrigine.66 cADRs represent a significant burden on the healthcare system, and SCAR with its most severe form, TEN, still carries a mortality rate as high as 35%.67 These life-threatening conditions could not be predicted and prevented until the recent advancement of pharmacogenomics in discovering of a strong association between a HLA allele (HLA-B*15:02) and carbamazepine-induced SJS/TEN in Han Chinese residing in Taiwan; these results were subsequently replicated in different populations (Figure 2).136869707172 The strong genetic association suggests a direct involvement of HLA in the pathogenesis of drug hypersensitivity. It has been shown that the HLA molecule presents an antigenic drug and results in clonal expansion and activation of CD8+cytotoxic T cells. A pharmacogenomic study also identified an unusual form of granulysin secreted by these cytotoxic T lymphocytes and natural killer cells responsible for the rapid and disseminated keratinocyte death in SJS/TEN.73 The high sensitivity and specificity of genetic markers provides a plausible basis for developing tests to identify individuals at risk for drug hypersensitivity. A large prospective study has shown that HLA-B*15:02screening before carbamazepine treatment can effectively reduce the incidence of carbamazepine-induced SJS/TEN.7475 Translational research demonstrates that preventing drug toxicity by screening people at risk before prescription of a drug is a clinical reality.75 Application of HLA-B*15:02genotyping as a screening tool for high risk populations taking carbamazepine is now recommended by many regulatory agencies worldwide, including Taiwan, USA, Thailand, Hong-Kong, and Singapore.

Figure 2.
Download full-size image
Figure 2. Severe adverse drug reactions with human leukocyte antigen (HLA) association. The interaction of HLA–T-cell receptor (TCR) drugs is the essential event for some types of severe adverse drug reactions. Some polymorphisms of genetic variants (e.g., HLA-B*15:02) related to this critical event could be used in clinical practice. APCs = antigen presenting cells.

This genetic association, albeit weaker, has also been found with phenytoin and lamotrigine (Table 3).7677 In Caucasian and Japanese populations the risk of developing hypersensitivity reactions to carbamazepine was found to be associated with another allele, HLA-A*31:01.7879 In addition to HLA, carbamazepine-induced SJS/TEN, a common type of AED-induced SCARs, was found to be associated with a specific T-cell receptor (Figure 2) clonotypes,80which could be another supportive biomarker to enhance the positive predictive value.81

4.2. Clozapine: HLA association of clozapine-induced agranulocytosis

Clozapine is a second-generation antipsychotic agent that belongs to the benzodiazepine group. It is indicated for the treatment of resistant schizophrenia or schizoaffective disorder and other psychotic conditions. Although clozapine has several advantages over conventional antipsychotics, the risk of agranulocytosis restricts its use. Based on a retrospective study, clozapine-induced agranulocytosis is associated with the rare HLA-DQB1(126Q) and HLA-B (158T) alleles. The pharmacogenetic test using these two polymorphisms has a sensitivity of 36% and specificity of 89% (Table 3).82 In addition, clozapine FDA label recommends for some CYP2D6 genotype-carrying patients a lower dose of clozapine (Table 3). However, a prospective study demonstrating whether a pharmacogenetic test based on this genetic association could have clinical utility and prevent clozapine-induced agranulocytosis has not yet been done.

4.3. Moving pharmacogenomic findings into medical practice for pain therapy

Before developing pharmacogenetic tests, a trial-and-error approach for analgesics was used for decades in analgesic treatment. Available pharmacogenomics studies indicated that some of the genetic markers have potential to be used clinically to guide the physicians to choose a right analgesic drug with a right dose to a right person. Some of the examples are listed in  These are based on well-defined genetic variants which have been replicated by others and have high predictive values (either high negative predictive value or high positive predictive value or both). These tests also have high sensitivity and specificity. Some of the tests have also been validated by a prospective clinical trial and already in routine medical practice; for example, HLA-B*15:02 screening to prevent carbamazepine-induced SJS.75Many of the tests also have clinical guidelines for dose adjustment and alternative medications assembled by The Clinical Pharmacogenomics Implementation Consortium (e.g., codeine and carbamazepine). Although there have been clear advances in clinical implementation of pharmacogenetics, some barriers slow down the speed of pharmacogenetic testing widespread.83 For example, long turnaround time of genetic tests, less value of genomic technologies after health economic evaluation, uncertain interpretation for the pharmacogenetic genetic test results, population restriction, and lack of prospective studies documenting superiority of the pharmacogenetic-guided treatment approach have to be overcome in the future of personalized medicine of pain.

5. Special consideration for pain medication: drug–drug interaction

“Drug–drug interaction,” defined as an interaction between two or more drugs, could alter the effectiveness, toxicity, or half-life of the interacting drugs. Moreover, it may increase the risk for serious ADR. Drug–drug interaction has been estimated to contribute to approximately 6–10% of ADRs.84 For pain management, more than one analgesic drug may be administered. In addition, for treating other diseases at the same time, such as coronary artery disease, hypertension, diabetes mellitus, and infections, multiple drugs may be also administered.85 The CYP450 enzyme pathway has been thought to be primarily involved in the metabolism of drugs, especially certain opioids used for the management of severe chronic pain. Therefore, the events underlying drug–drug interactions in pain management using multiple drugs could explain why pharmacogenetic tests for the CYP gene are helpful for identification of the optimal drug dose and prevention of ADRs. Furthermore, some information regarding drug–drug interactions could be referred to on drug labels before administration of analgesic drugs. For example, the interaction between quetiapine (for the relief of pain or depression) and ritonavir (for the treatment of human immunodeficiency virus infection) may lead to rapid and severe weight gain.86

6. Summary and future directions

Despite the pharmacogenomic studies that show that genetic variants may account for individual differences in drug responses, few genetic markers can be used clinically to guide the dosage for optimal efficacy of pain therapies.8There are several reasons for the slow progress in this regard. Firstly, the pain sensitivity varies for individual patients and underlying causes of pain are not well characterized. The heterogeneous conditions could interfere with the evaluation of drug efficacy. More precise phenotypes are needed.87 Secondly, the prior usage of opioid and tolerance may affect the drug response. Thirdly, multiple genes may affect drug pharmacokinetics and pharmacodynamics and there are also drug–drug interactions, which may complicate the genetic studies. Fourthly, because the drugs used in pain management alter multiple signaling pathways, and the efficacy of drugs could not be easily measured due to the lack of key indicators or outcome. Therefore, pain management with drugs such as morphine using a “titration” method is currently still widely used in the clinic to guide dosage decisions.8889 By contrast, clinically useful genetic variants for ADRs are easier to identify based on pharmacogenomic studies, because some ADR conditions have clear phenotypes (i.e., SJS or TEN), critical pathways (i.e., HLA–T-cell receptor; Table 3 and Figure 2),80 and specific indicators (i.e., granulysin).73

In conclusion, the clinically useful pharmacogenomic tests currently available for pain therapy are more direct toward at predicting drug toxicity or dose adjustment. More studies are needed to identify genetic variants that determine drug efficacy of pain medications. This area of research is likely rapidly accelerating with the reducing cost of next-generation sequencing and well-established biobanking system worldwide.

Acknowledgments

We gratefully acknowledge the members of the Translational Resource Center and the National Center for Genome Medicine at Academia Sinica for their support. This study was supported by the Academia Sinica Genomic Medicine Multicenter Study (40-05-GMM). The funders had no role in study design, data collection, or analysis, the decision to publish or preparation of the manuscript.


References

1
F.S. Collins, H. Varmus
A new initiative on precision medicine
N Engl J Med, 372 (2015), pp. 793-795
2
E.A. Ashley
The precision medicine initiative: a new national effort
JAMA, 313 (2015), pp. 2119-2120
3
S.S. Ladak, V.W. Chan, T. Easty, A. Chagpar
Right medication, right dose, right patient, right time, and right route: how do we select the right patient-controlled analgesia (PCA) device?
Pain Manag Nurs, 8 (2007), pp. 140-145
4
C. Ono, H. Kikkawa, A. Suzuki, M. Suzuki, Y. Yamamoto, K. Ichikawa, et al.
Clinical impact of genetic variants of drug transporters in different ethnic groups within and across regions
Pharmacogenomics, 14 (2013), pp. 1745-1764
5
C.M. Flores, J.S. Mogil
The pharmacogenetics of analgesia: toward a genetically-based approach to pain management
Pharmacogenomics, 2 (2001), pp. 177-194
6
C.Y. Wei, M.T. Lee, Y.T. Chen
Pharmacogenomics of adverse drug reactions: implementing personalized medicine
Hum Mol Genet, 21 (2012), pp. R58-R65
7
A.H. Maitland-van der Zee, A. Alfirevic
Pharmacogenetics and pharmacogenomics: practical applications in routine medical practice
Genome Med, 2 (2010), p. 69
8
M. Pirmohamed
Personalized pharmacogenomics: predicting efficacy and adverse drug reactions
Annu Rev Genomics Hum Genet, 15 (2014), pp. 349-370
9
A.R. Jadad, G.P. Browman
The WHO analgesic ladder for cancer pain management. Stepping up the quality of its evaluation
JAMA, 274 (1995), pp. 1870-1873
10
T. Rabben, I. Oye
Interindividual differences in the analgesic response to ketamine in chronic orofacial pain
Eur J Pain, 5 (2001), pp. 233-240
11
S. Sadhasivam, V. Chidambaran, V.A. Olbrecht, H.R. Esslinger, K. Zhang, X. Zhang, et al.
Genetics of pain perception, COMT and postoperative pain management in children
Pharmacogenomics, 15 (2014), pp. 277-284
12
E. Lemoli, M.C. Vivirito, M. Coen, I. Faggion, T. Quirino
Anticonvulsant hypersensitivity syndrome due to carbamazepine
Allergy, 54 (1999), pp. 1329-1330
Article  
13
W.H. Chung, S.I. Hung, H.S. Hong, M.S. Hsih, L.C. Yang, H.C. Ho, et al.
Medical genetics: a marker for Stevens–Johnson syndrome
Nature, 428 (2004), p. 486
14
H. Knotkova, M. Pappagallo
Adjuvant analgesics
Med Clin North Am, 91 (2007), pp. 113-124
15
V.K. Kong, M.G. Irwin
Adjuvant analgesics in neuropathic pain
Eur J Anaesthesiol, 26 (2009), pp. 96-100
16
D. Nishizawa, M. Nagashima, R. Katoh, Y. Satoh, M. Tagami, S. Kasai, et al.
Association between KCNJ6 (GIRK2) gene polymorphisms and postoperative analgesic requirements after major abdominal surgery
PloS One, 4 (2009), p. e7060
Article   CrossRef  
17
S. Ide, D. Nishizawa, K. Fukuda, S. Kasai, J. Hasegawa, M. Hayashida, et al.
Haplotypes of P2RX7 gene polymorphisms are associated with both cold pain sensitivity and analgesic effect of fentanyl
Mol Pain, 10 (2014), p. 75
Article  
18
M. Hayashida, M. Nagashima, Y. Satoh, R. Katoh, M. Tagami, S. Ide, et al.
Analgesic requirements after major abdominal surgery are associated with OPRM1 gene polymorphism genotype and haplotype
Pharmacogenomics, 9 (2008), pp. 1605-1616
19
M. Ingelman-Sundberg, M. Oscarson, R.A. McLellan
Polymorphic human cytochrome P450 enzymes: an opportunity for individualized drug treatment
Trends Pharmacol Sci, 20 (1999), pp. 342-349
20
U.M. Stamer, K. Lehnen, F. Hothker, B. Bayerer, S. Wolf, A. Hoeft, et al.
Impact of CYP2D6 genotype on postoperative tramadol analgesia
Pain, 105 (2003), pp. 231-238
21
P. Madadi, D. Avard, G. Koren
Pharmacogenetics of opioids for the treatment of acute maternal pain during pregnancy and lactation
Curr Drug Metab, 13 (2012), pp. 721-727
22
W. Leppert
CYP2D6 in the metabolism of opioids for mild to moderate pain
Pharmacology, 87 (2011), pp. 274-285
23
A.A. Somogyi, D.T. Barratt, J.K. Coller
Pharmacogenetics of opioids
Clin Pharmacol Ther, 81 (2007), pp. 429-444
24
B.H. McCarberg, R.L. Barkin
Long-acting opioids for chronic pain: pharmacotherapeutic opportunities to enhance compliance, quality of life, and analgesia
Am J Ther, 8 (2001), pp. 181-186
25
B.R. Overholser, D.R. Foster
Opioid pharmacokinetic drug–drug interactions
Am J Manag Care, 17 (2011), pp. S276-S287
26
E. Chevlen
Opioids: a review
Curr Pain Headache Rep, 7 (2003), pp. 15-23
27
R.F. Rodriguez, J.M. Castillo, M. Del Pilar Castillo, P.D. Nunez, M.F. Rodriguez, J.M. Restrepo, et al.
Codeine/acetaminophen and hydrocodone/acetaminophen combination tablets for the management of chronic cancer pain in adults: a 23-day, prospective, double-blind, randomized, parallel-group study
Clin Ther, 29 (2007), pp. 581-587
28
R. Wolford, J. Kahler, P. Mishra, P. Vasilenko, R. DeYoung
A prospective comparison of transnasal butorphanol and acetaminophen with codeine for the relief of acute musculoskeletal pain
Am J Emerg Med, 15 (1997), pp. 101-103
29
H.V. Pai, R.P. Kommaddi, S.J. Chinta, T. Mori, M.R. Boyd, V. Ravindranath
A frameshift mutation and alternate splicing in human brain generate a functional form of the pseudogene cytochrome P4502D7 that demethylates codeine to morphine
J Biol Chem, 279 (2004), pp. 27383-27389
30
P.A. Crooks, S.G. Kottayil, A.M. Al-Ghananeem, S.R. Byrn, D.A. Butterfield
Opiate receptor binding properties of morphine-, dihydromorphine-, and codeine 6-O-sulfate ester congeners
Bioorg Med Chem Lett, 16 (2006), pp. 4291-4295
31
Z.R. Chen, A.A. Somogyi, F. Bochner
Polymorphic O-demethylation of codeine
Lancet, 2 (1988), pp. 914-915
32
L.C. Fernandes, T. Kilicarslan, H.L. Kaplan, R.F. Tyndale, E.M. Sellers, M.K. Romach
Treatment of codeine dependence with inhibitors of cytochrome P450 2D6
J Clin Psychopharmacol, 22 (2002), pp. 326-329
33
K.R. Crews, A. Gaedigk, H.M. Dunnenberger, T.E. Klein, D.D. Shen, J.T. Callaghan, et al.
Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for codeine therapy in the context of cytochrome P450 2D6 (CYP2D6) genotype
Clin Pharmacol Ther, 91 (2012), pp. 321-326
34
Z. Zahari, R. Ismail
Influence of Cytochrome P450, Family 2, Subfamily D, Polypeptide 6 (CYP2D6) polymorphisms on pain sensitivity and clinical response to weak opioid analgesics
Drug Metab Pharmacokinet, 29 (2014), pp. 29-43
35
M.T. Susce, E. Murray-Carmichael, J. de Leon
Response to hydrocodone, codeine and oxycodone in a CYP2D6 poor metabolizer
Prog Neuropsychopharmacol Biol Psychiatry, 30 (2006), pp. 1356-1358
36
J.K. Coller, C. Joergensen, D.J. Foster, H. James, D. Gillis, L. Christrup, et al.
Lack of influence of CYP2D6 genotype on the clearance of (R)-, (S)- and racemic-methadone
Int J Clin Pharmacol Ther, 45 (2007), pp. 410-417
37
M. King, W. Su, A. Chang, A. Zuckerman, G.W. Pasternak
Transport of opioids from the brain to the periphery by P-glycoprotein: peripheral actions of central drugs
Nat Neurosci, 4 (2001), pp. 268-274
38
H.E. Hassan, S.L. Mercer, C.W. Cunningham, A. Coop, N.D. Eddington
Evaluation of the P-glycoprotein (Abcb1) affinity status of a series of morphine analogs: comparative study with meperidine analogs to identify opioids with minimal P-glycoprotein interactions
Int J Pharm, 375 (2009), pp. 48-54
39
J.E. Robinson, E. Vardy, J.F. DiBerto, V.I. Chefer, K.L. White, E.W. Fish, et al.
Receptor reserve moderates mesolimbic responses to opioids in a humanized mouse model of the OPRM1 A118G polymorphism
Neuropsychopharmacology, 40 (2015), pp. 2614-2622
40
A. Behrooz
Pharmacogenetics and anaesthetic drugs: Implications for perioperative practice
Ann Med Surg (Lond), 4 (2015), pp. 470-474
41
S.M. Spampinato
Overview of genetic analysis of human opioid receptors
Methods Mol Biol, 1230 (2015), pp. 3-12
42
T. Kroslak, K.S. Laforge, R.J. Gianotti, A. Ho, D.A. Nielsen, M.J. Kreek
The single nucleotide polymorphism A118G alters functional properties of the human mu opioid receptor
J Neurochem, 103 (2007), pp. 77-87
43
J.T. Payte
A brief history of methadone in the treatment of opioid dependence: a personal perspective
J Psychoactive Drugs, 23 (1991), pp. 103-107
Article   CrossRef  
44
E. Bruera, C. Sweeney
Methadone use in cancer patients with pain: a review
J Palliat Med, 5 (2002), pp. 127-138
45
A. Srivastava, M. Kahan
Methadone induction doses: are our current practices safe?
J Addict Dis, 25 (2006), pp. 5-13
46
Y. Li, J.P. Kantelip, P. Gerritsen-van Schieveen, S. Davani
Interindividual variability of methadone response: impact of genetic polymorphism
Mol Diagn Ther, 12 (2008), pp. 109-124
47
B.A. Chizh, H. Schlutz, M. Scheede, W. Englberger
The N-methyl-D-aspartate antagonistic and opioid components of d-methadone antinociception in the rat spinal cord
Neurosci Lett, 296 (2000), pp. 117-120
48
D. Moulin
Use of methadone for neuropathic pain
Pain Res Manag, 8 (2003), pp. 131-132
49
M.L. Sotgiu, M. Valente, R. Storchi, G. Caramenti, G.E. Biella
Cooperative N-methyl-D-aspartate (NMDA) receptor antagonism and mu-opioid receptor agonism mediate the methadone inhibition of the spinal neuron pain-related hyperactivity in a rat model of neuropathi
Pharmacol Res, 60 (2009), pp. 284-290
50
A.L. Gorman, K.J. Elliott, C.E. Inturrisi
The d- and l-isomers of methadone bind to the non-competitive site on the N-methyl-D-aspartate (NMDA) receptor in rat forebrain and spinal cord
Neurosci Lett, 223 (1997), pp. 5-8
51
Health Quality Ontario
Optimum methadone compliance testing: an evidence-based analysis
Ont Health Technol Assess Ser, 6 (2006), pp. 1-54
52
C.B. Eap, T. Buclin, P. Baumann
Interindividual variability of the clinical pharmacokinetics of methadone: implications for the treatment of opioid dependence
Clin Pharmacokinet, 41 (2002), pp. 1153-1193
53
J.A. Agundez, C. Martinez, E. Garcia-Martin, J.M. Ladero
Cytochrome P450 CYP2C9 polymorphism and NSAID-related acute gastrointestinal bleeding
Gastroenterology, 133 (2007), pp. 2071-2072 author reply 72–3
54
C. Jaja, N. Patel, S.A. Scott, R. Gibson, A. Kutlar
CYP2C9 allelic variants and frequencies in a pediatric sickle cell disease cohort: implications for NSAIDs pharmacotherapy
Clin Transl Sci, 7 (2014), pp. 396-401
55
J.B. Dahl, R.V. Nielsen, J. Wetterslev, L. Nikolajsen, K. Hamunen, V.K. Kontinen, et al.
Post-operative analgesic effects of paracetamol, NSAIDs, glucocorticoids, gabapentinoids and their combinations: a topical review
Acta Anaesthesiol Scand, 58 (2014), pp. 1165-1181
56
E.T. Mellerup, P. Plenge
Specific binding sites for cyclic antidepressants
Acta Pharmacol Toxicol (Copenh), 56 (1985), pp. 154-161
57
E. Pallis, A. Vasilaki, D. Fehlmann, A. Kastellakis, D. Hoyer, C. Spyraki, et al.
Antidepressants influence somatostatin levels and receptor pharmacology in brain
Neuropsychopharmacology, 34 (2009), pp. 952-963
58
E. Spina, M.G. Scordo
Clinically significant drug interactions with antidepressants in the elderly
Drugs Aging, 19 (2002), pp. 299-320
59
A. Seeringer, J. Kirchheiner
Pharmacogenetics-guided dose modifications of antidepressants
Clin Lab Med, 28 (2008), pp. 619-626
60
A. Vadalouca, E. Raptis, E. Moka, P. Zis, P. Sykioti, I. Siafaka
Pharmacological treatment of neuropathic cancer pain: a comprehensive review of the current literature
Pain Pract, 12 (2012), pp. 219-251
61
J. Kirchheiner, K. Brosen, M.L. Dahl, L.F. Gram, S. Kasper, I. Roots, et al.
CYP2D6 and CYP2C19 genotype-based dose recommendations for antidepressants: a first step towards subpopulation-specific dosages
Acta Psychiatr Scand, 104 (2001), pp. 173-192
62
J. Kirchheiner, K. Nickchen, M. Bauer, M.L. Wong, J. Licinio, I. Roots, et al.
Pharmacogenetics of antidepressants and antipsychotics: the contribution of allelic variations to the phenotype of drug response
Mol Psychiatry, 9 (2004), pp. 442-473
63
T. Rau, G. Wohlleben, H. Wuttke, N. Thuerauf, J. Lunkenheimer, M. Lanczik, et al.
CYP2D6 genotype: impact on adverse effects and nonresponse during treatment with antidepressants-a pilot study
Clin Pharmacol Ther, 75 (2004), pp. 386-393
64
D.K. Chang, N.H. Shear
Cutaneous reactions to anticonvulsants
Semin Neurol, 12 (1992), pp. 329-337
65
J.C. Roujeau, J.P. Kelly, L. Naldi, B. Rzany, R.S. Stern, T. Anderson, et al.
Medication use and the risk of Stevens–Johnson syndrome or toxic epidermal necrolysis
N Engl J Med, 333 (1995), pp. 1600-1607
66
L.E. French
Toxic epidermal necrolysis and Stevens–Johnson syndrome: our current understanding
Allergol Int, 55 (2006), pp. 9-16
67
T.Y. Mehta, L.M. Prajapati, B. Mittal, C.G. Joshi, J.J. Sheth, D.B. Patel, et al.
Association of HLA-B*1502 allele and carbamazepine-induced Stevens–Johnson syndrome among Indians
Indian J Dermatol, Venereol Leprol, 75 (2009), pp. 579-582
68
C. Locharernkul, J. Loplumlert, C. Limotai, W. Korkij, T. Desudchit, S. Tongkobpetch, et al.
Carbamazepine and phenytoin induced Stevens–Johnson syndrome is associated with HLA-B*1502 allele in Thai population
Epilepsia, 49 (2008), pp. 2087-2091
69
S.H. Kim, K.W. Lee, W.J. Song, S.H. Kim, Y.K. Jee, S.M. Lee, et al.
Carbamazepine-induced severe cutaneous adverse reactions and HLA genotypes in Koreans
Epilepsy Res, 97 (2011), pp. 190-197
70
S.M. Then, Z.Z. Rani, A.A. Raymond, S. Ratnaningrum, R. Jamal
Frequency of the HLA-B*1502 allele contributing to carbamazepine-induced hypersensitivity reactions in a cohort of Malaysian epilepsy patients
Asian Pac J Allergy Immunol, 29 (2011), pp. 290-293
71
D.V. Nguyen, H.C. Chu, D.V. Nguyen, M.H. Phan, T. Craig, K. Baumgart, et al.
HLA-B*1502 and carbamazepine-induced severe cutaneous adverse drug reactions in Vietnamese
Asia Pac Allergy, 5 (2015), pp. 68-77
72
W.H. Chung, S.I. Hung, J.Y. Yang, S.C. Su, S.P. Huang, C.Y. Wei, et al.
Granulysin is a key mediator for disseminated keratinocyte death in Stevens–Johnson syndrome and toxic epidermal necrolysis
Nat Med, 14 (2008), pp. 1343-1350
73
Z. Chen, D. Liew, P. Kwan
Real-world efficiency of pharmacogenetic screening for carbamazepine-induced severe cutaneous adverse reactions
PLoS One, 9 (2014), p. e96990
CrossRef  
74
P. Chen, J.J. Lin, C.S. Lu, C.T. Ong, P.F. Hsieh, C.C. Yang, et al.
Carbamazepine-induced toxic effects and HLA-B*1502 screening in Taiwan
N Engl J Med, 364 (2011), pp. 1126-1133
75
S.I. Hung, W.H. Chung, Z.S. Liu, C.H. Chen, M.S. Hsih, R.C. Hui, et al.
Common risk allele in aromatic antiepileptic-drug induced Stevens–Johnson syndrome and toxic epidermal necrolysis in Han Chinese
Pharmacogenomics, 11 (2010), pp. 349-356
76
X. Li, K. Yu, S. Mei, J. Huo, J. Wang, Y. Zhu, et al.
HLA-B*1502 increases the risk of phenytoin or lamotrigine induced Stevens–Johnson syndrome/toxic epidermal necrolysis: evidence from a meta-analysis of nine case-control studies
Drug Res, 65 (2015), pp. 107-111
77
M. McCormack, A. Alfirevic, S. Bourgeois, J.J. Farrell, D. Kasperaviciute, M. Carrington, et al.
HLA-A*3101 and carbamazepine-induced hypersensitivity reactions in Europeans
N Engl J Med, 364 (2011), pp. 1134-1143
78
T. Ozeki, T. Mushiroda, A. Yowang, A. Takahashi, M. Kubo, Y. Shirakata, et al.
Genome-wide association study identifies HLA-A*3101 allele as a genetic risk factor for carbamazepine-induced cutaneous adverse drug reactions in Japanese population
Hum Mol Genet, 20 (2011), pp. 1034-1041
79
T.M. Ko, W.H. Chung, C.Y. Wei, H.Y. Shih, J.K. Chen, C.H. Lin, et al.
Shared and restricted T-cell receptor use is crucial for carbamazepine-induced Stevens–Johnson syndrome
J Allergy Clin Immunol, 128 (2011), pp. 1266-1276
80
T.M. Ko, Y.T. Chen
T-cell receptor and carbamazepine-induced Stevens–Johnson syndrome and toxic epidermal necrolysis: understanding a hypersensitivity reaction
Expert Rev Clin Immunol, 8 (2012), pp. 467-477
81
J.I. Goldstein, L.F. Jarskog, C. Hilliard, A. Alfirevic, L. Duncan, D. Fourches, et al.
Clozapine-induced agranulocytosis is associated with rare HLA-DQB1 and HLA-B alleles
Nat Commun, 5 (2014), p. 4757
Article  
82
M.V. Relling, W.E. Evans
Pharmacogenomics in the clinic
Nature, 526 (2015), pp. 343-350
83
W.N. Kelly
Potential risks and prevention, Part 2: drug-induced permanent disabilities
Am J Health Syst Pharm, 58 (2001), pp. 1325-1329
84
M. Marwan, T. Pflederer, T. Schepis, M. Seltmann, L. Klinghammer, G. Muschiol, et al.
Accuracy of dual-source CT to identify significant coronary artery disease in patients with uncontrolled hypertension presenting with chest pain: comparison with coronary angiography
Int J Cardiovasc Imaging, 28 (2012), pp. 1173-1180
85
T.M. Pollack, C. McCoy, W. Stead
Clinically significant adverse events from a drug interaction between quetiapine and atazanavir–ritonavir in two patients
Pharmacotherapy, 29 (2009), pp. 1386-1391
86
K. Dunleavy, M. Roschewski, W.H. Wilson
Precision treatment of distinct molecular subtypes of diffuse large B-cell lymphoma: ascribing treatment based on the molecular phenotype
Clin Cancer Res, 20 (2014), pp. 5182-5193
87
F. Aubrun, S. Monsel, O. Langeron, P. Coriat, B. Riou
Postoperative titration of intravenous morphine
Eur J Anaesthesiol, 18 (2001), pp. 159-165
88
F. Aubrun, J.X. Mazoit, B. Riou
Postoperative intravenous morphine titration
Br J Anaesth, 108 (2012), pp. 193-201

References

Close