51
|
Abstract
Abstract
Background:
Although a number of studies have investigated the association of the OPRM1 A118G polymorphism with pain response, a consensus has not yet been reached.
Methods:
The authors searched PubMed, EMBASE, and the Cochrane Library to identify gene-association studies that explored the impact of the OPRM1 A118G polymorphism on postoperative opioid requirements through July 2013. Two evaluators independently reviewed and selected articles on the basis of prespecified selection criteria. The authors primarily investigated the standardized mean difference (SMD) of required amounts of opioids between AA homozygotes and G-allele carriers. The authors also performed subgroup analyses for race, opioid use, and type of surgery. Potential bias was assessed using the Egger’s test with a trim and fill procedure.
Results:
Three hundred forty-six articles were retrieved from databases, and 18 studies involving 4,607 participants were included in the final analyses. In a random-effect meta-analysis, G-allele carriers required a higher mean opioid dose than AA homozygotes (SMD, −0.18; P = 0.003). Although there was no evidence of publication bias, heterogeneity was present among studies (I2 = 66.8%). In the subgroup meta-analyses, significance remained robust in Asian patients (SMD, −0.21; P = 0.001), morphine users (SMD, −0.29; P <0.001), and patients who received surgery for a viscus (SMD, −0.20; P = 0.008).
Conclusions:
The OPRM1 A118G polymorphism was associated with interindividual variability in postoperative response to opioids. In a subpopulation, identifying OPRM1 A118G polymorphism may provide valuable information regarding the individual analgesic doses that are required to achieve satisfactory pain control.
Collapse
|
52
|
Abstract
Postoperative nausea and vomiting (PONV) is a long-standing issue, not a new concept in anesthesiology. Despite many studies over the last several decades, PONV remains a significant problem due to its complex mechanism. This review presents a summary of the mechanism underlying the pathogenesis of PONV, focusing on preventive treatment, particularly the use of new drugs. In addition, we discuss the latest meta-analysis results regarding correct clinical use of classic drugs. I also summarize the latest trends of postdischarge nausea and vomiting and the pharmacogenetics, which is attracting a great deal of attention from other medical fields in PONV-related studies. Finally, we discuss the drawbacks of existing studies on PONV and suggest a focus for future investigations.
Collapse
Affiliation(s)
- Young Eun Moon
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary's Hospital, Catholic University College of Medicine, Seoul, Korea
| |
Collapse
|
53
|
Nielsen LM, Olesen AE, Branford R, Christrup LL, Sato H, Drewes AM. Association Between Human Pain-Related Genotypes and Variability in Opioid Analgesia: An Updated Review. Pain Pract 2014; 15:580-94. [DOI: 10.1111/papr.12232] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 06/04/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Lecia M. Nielsen
- Mech-Sense; Department of Gastroenterology and Hepatology; Aalborg University Hospital; Aalborg Denmark
- Department of Drug Design and Pharmacology; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - Anne E. Olesen
- Mech-Sense; Department of Gastroenterology and Hepatology; Aalborg University Hospital; Aalborg Denmark
| | - Ruth Branford
- Department of Palliative Medicine; Royal Marsden Hospital; London UK
| | - Lona L. Christrup
- Department of Drug Design and Pharmacology; Faculty of Health and Medical Sciences; University of Copenhagen; Copenhagen Denmark
| | - Hiroe Sato
- Interstitial Lung Disease Unit; Royal Brompton Hospital & National Heart and Lung Institute; Imperial College London; London UK
| | - Asbjørn M. Drewes
- Mech-Sense; Department of Gastroenterology and Hepatology; Aalborg University Hospital; Aalborg Denmark
- Department of Clinical Medicine; Aalborg University; Aalborg Denmark
| |
Collapse
|
54
|
Association of μ-opioid receptor gene (OPRM1) haplotypes with postoperative nausea and vomiting. Exp Brain Res 2014; 232:2627-35. [DOI: 10.1007/s00221-014-3987-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 05/06/2014] [Indexed: 12/31/2022]
|
55
|
Genetic factors associated with pharmacotherapy and background sensitivity to postoperative and chemotherapy-induced nausea and vomiting. Exp Brain Res 2014; 232:2613-25. [PMID: 24792505 DOI: 10.1007/s00221-014-3968-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/03/2014] [Indexed: 12/12/2022]
Abstract
Postoperative nausea and vomiting (PONV) continues to be a most common complication of surgery and anesthesia. It has been suggested that the inherited factors may play a significant role in the background sensitivity to both PONV and also chemotherapy-induced nausea and vomiting (CINV), including resistance to antiemetic prophylaxis and/or therapy. This notion could be best exemplified by occurrence of PONV in several generations of families and concordance of PONV in monozygotic twins. The most frequently addressed issue in the research on genomic background of PONV/CINV relates to the inherited resistance to the antiemetic treatment (pharmacogenomics), and in lesser degree to their genomic background. The most common group of antiemetics consists of 5HT3 receptor antagonists, and this group was an initial target of pharmacogenomic research. Most research approaches have been based on the investigation of polymorphic variations in the target for the antiemetic 5HT3 receptor antagonists, i.e., serotonin receptor subunits A and B (HTR3A and HTR3B). The other area of pharmacogenomic investigations includes metabolic pathways of 5HT3 antagonists, in particular polymorphic variants of the CYP450 2D6 isoform (CYP2D6) because most of them are metabolized in various degrees by the CYP2D6 system. The results of targeted genomic association studies indicate that other genes are also associated with PONV and CINV, including OPRM1, and ABCB1. In addition, genes such as DRD2 and CHRM3 genes have recently been associated with PONV. The new genome-wide association studies seem also to indicate that the background genomic sensitivity to PONV and CINV might be multifactorial and include several genomic pathways.
Collapse
|
56
|
Opioid analgesics and narcotic antagonists. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/b978-0-444-62635-6.00008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
|
57
|
Anand KJS. Pediatric critical care: grand challenges for a glowing future. Front Pediatr 2014; 2:35. [PMID: 24818121 PMCID: PMC4012217 DOI: 10.3389/fped.2014.00035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 04/16/2014] [Indexed: 01/19/2023] Open
Affiliation(s)
- Kanwaljeet J S Anand
- Pain Neurobiology Laboratory, Department of Pediatrics, University of Tennessee Neuroscience Institute, University of Tennessee Health Science Center , Memphis, TN , USA
| |
Collapse
|
58
|
Horn CC, Meyers K, Oberlies N. Musk shrews selectively bred for motion sickness display increased anesthesia-induced vomiting. Physiol Behav 2013; 124:129-37. [PMID: 24239993 DOI: 10.1016/j.physbeh.2013.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 11/06/2013] [Indexed: 10/26/2022]
Abstract
Susceptibility to motion sickness is a predictor of postoperative nausea and vomiting, and studies in humans suggest that genetic factors determine sensitivity to motion sickness. The aim of the current study was to determine if a preclinical model could be selectively bred for motion-induced emesis and to assess a potential relationship to anesthesia-induced emesis. Musk shrews were tested for motion-induced emesis using a shaker plate (10min, 1Hz, and 4cm of lateral displacement). Animals were rank ordered for motion-induced emesis and selectively bred to produce high and low response strains. Shrews were also tested with nicotine (5mg/kg, sc), copper sulfate (CuSO4; 120mg/kg, ig), and isoflurane anesthesia (10min; 3%) to measure responses to a panel of emetic stimuli. High response strain shrews demonstrated significantly more emetic episodes to motion exposure compared to low response strain animals in the F1 and F2 generations. In F2 animals, there were no significant differences in total emetic responses or emetic latency between strains after nicotine injection or CuSO4 gavage. However, isoflurane exposure stimulated more emesis in F1 and F2 high versus low strain animals, which suggests a relationship between vestibular- and inhalational anesthesia-induced emesis. Overall, these results indicate genetic determinants of motion sickness in a preclinical model and a potential common mechanism for motion sickness and inhalational anesthesia-induced emesis. Future work may include genetic mapping of potential "emetic sensitivity genes" to develop novel therapies or diagnostics for patients with high risk of nausea and vomiting.
Collapse
Affiliation(s)
- Charles C Horn
- Biobehavioral Medicine in Oncology Program, Univ. of Pittsburgh Cancer Institute, Pittsburgh, PA, USA; Dept. of Medicine, Div. of Gastroenterology, Hepatology, and Nutrition, Univ. of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Dept. of Anesthesiology, Univ. of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Neuroscience, Univ. of Pittsburgh, Pittsburgh, PA, USA.
| | | | | |
Collapse
|
59
|
Horn CC, Wallisch WJ, Homanics GE, Williams JP. Pathophysiological and neurochemical mechanisms of postoperative nausea and vomiting. Eur J Pharmacol 2013; 722:55-66. [PMID: 24495419 DOI: 10.1016/j.ejphar.2013.10.037] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 10/07/2013] [Accepted: 10/08/2013] [Indexed: 12/13/2022]
Abstract
Clinical research shows that postoperative nausea and vomiting (PONV) is caused primarily by the use of inhalational anesthesia and opioid analgesics. PONV is also increased by several risk predictors, including a young age, female sex, lack of smoking, and a history of motion sickness. Genetic studies are beginning to shed light on the variability in patient experiences of PONV by assessing polymorphisms of gene targets known to play roles in emesis (serotonin type 3, 5-HT3; opioid; muscarinic; and dopamine type 2, D2, receptors) and the metabolism of antiemetic drugs (e.g., ondansetron). Significant numbers of clinical trials have produced valuable information on pharmacological targets important for controlling PONV (e.g., 5-HT3 and D2), leading to the current multi-modal approach to inhibit multiple sites in this complex neural system. Despite these significant advances, there is still a lack of fundamental knowledge of the mechanisms that drive the hindbrain central pattern generator (emesis) and forebrain pathways (nausea) that produce PONV, particularly the responses to inhalational anesthesia. This gap in knowledge has limited the development of novel effective therapies of PONV. The current review presents the state of knowledge on the biological mechanisms responsible for PONV, summarizing both preclinical and clinical evidence. Finally, potential ways to advance the research of PONV and more recent developments on the study of postdischarge nausea and vomiting (PDNV) are discussed.
Collapse
Affiliation(s)
- Charles C Horn
- Biobehavioral Medicine in Oncology Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA; Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| | - William J Wallisch
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Gregg E Homanics
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - John P Williams
- Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
60
|
Smith HS, Laufer A. Opioid induced nausea and vomiting. Eur J Pharmacol 2013; 722:67-78. [PMID: 24157979 DOI: 10.1016/j.ejphar.2013.09.074] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 09/30/2013] [Accepted: 09/30/2013] [Indexed: 02/06/2023]
Abstract
Opioids are broad spectrum analgesics that are an integral part of the therapeutic armamentarium to combat pain in the palliative care population. Unfortunately, among the adverse effects of opioids that may be experienced along with analgesia is nausea, vomiting, and/or retching. Although it is conceivable that in the future, using combination agents (opioids combined with agents which may nullify emetic effects), currently nausea/vomiting remains a significant issue for certain patients. However, there exists potential current strategies that may be useful in efforts to diminish the frequency and/or intensity of opioid-induced nausea/vomiting (OINV).
Collapse
Affiliation(s)
- Howard S Smith
- Albany Medical College, Department of Anesthesiology, 47 New Scotland Avenue, MC-131, Albany, NY 12208, USA.
| | - Andras Laufer
- Albany Medical College, Department of Anesthesiology, 47 New Scotland Avenue, MC-131, Albany, NY 12208, USA.
| |
Collapse
|
61
|
Sia AT, Lim Y, Lim EC, Ocampo CE, Lim WY, Cheong P, Tan EC. Influence of Mu-Opioid Receptor Variant on Morphine Use and Self-Rated Pain Following Abdominal Hysterectomy. THE JOURNAL OF PAIN 2013; 14:1045-52. [DOI: 10.1016/j.jpain.2013.03.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Revised: 03/08/2013] [Accepted: 03/27/2013] [Indexed: 11/16/2022]
|
62
|
Xu Y, Johnson A. Opioid therapy pharmacogenomics for noncancer pain: efficacy, adverse events, and costs. PAIN RESEARCH AND TREATMENT 2013; 2013:943014. [PMID: 24167729 PMCID: PMC3791560 DOI: 10.1155/2013/943014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 08/18/2013] [Accepted: 08/18/2013] [Indexed: 01/16/2023]
Abstract
Chronic non-cancer pain is a debilitating condition associated with high individual and societal costs. While opioid treatment for pain has been available for centuries, it is associated with high variability in outcome, and a considerable proportion of patients is unable to attain relief from symptoms while suffering adverse events and developing medication dependence. We performed a review of the efficacy of pharmacogenomic markers and their abilities to predict adverse events, dependence, and associated economic costs, focusing on two genes: OPRM1 and CYP2D6. Data sources were articles indexed by PubMed on or before August 6, 2013. Articles were first selected after review of their titles and abstracts, and full papers were read to confirm eligibility. Initially, fifty-two articles were identified. Of these, 17 were relevant to biological actions of pharmacogenomic markers and their effect on therapeutic efficacy, 16 to adverse events, 15 to opioid dependence, and eight to economic costs. In conclusion, increasing costs of opioid therapy have made the advances in pharmacogenomics an attractive solution to personalize care with unclear repercussions related to the impact on costs, morbidity, and outcomes. This intersection of pharmacoeconomics and pharmacogenomics presents a unique platform to further examine current advances in clinical medicine and their utility in cost-effective treatment of chronic pain.
Collapse
Affiliation(s)
- Yan Xu
- Queen's University, Kingston, ON, Canada K7L 3N6
| | - Ana Johnson
- Department of Population Health Sciences, Queen's University, Kingston, ON, Canada K7L 3N6
- Institute for Clinical Evaluative Services, Queen's University, Kingston, ON, Canada K7L 3N6
| |
Collapse
|
63
|
Abstract
Studies on genetic contributions to labor analgesia have essentially evaluated the μ-opioid receptor gene (OPRM1), with some evidence that p.118A/G of OPRM1 influences the response to neuraxial opioids. As for labor progress, the β2-adrenergic receptor gene (ADRB2) is associated with preterm labor and delivery, and impacts the course of labor. Taken together though, there is no evidence that pharmacogenetic testing is needed or beneficial in the context of obstetric anesthesia; however, realizing the influence of genetic variants on specific phenotypes provides the rationale for a more cautious interpretation of clinical studies that attempt to find a dose-regimen that fits all.
Collapse
Affiliation(s)
- Ruth Landau
- Department of Anesthesiology and Pain Medicine, University of Washington Medical Center, 1959 NE Pacific Street, Seattle, WA 98195-6540, USA.
| |
Collapse
|
64
|
|
65
|
Sadhasivam S, Chidambaran V. Pharmacogenomics of opioids and perioperative pain management. Pharmacogenomics 2013; 13:1719-40. [PMID: 23171337 DOI: 10.2217/pgs.12.152] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Inadequate pain relief and adverse effects from analgesics remain common in children and adults during the perioperative period. Opioids are the most commonly used analgesics in children and adults to treat perioperative pain. Narrow therapeutic index and a large interpatient variability in response to opioids are clinically significant, with inadequate pain relief at one end of the spectrum and serious side effects, such as respiratory depression and excessive sedation due to relative overdosing, at the other end. Personalizing analgesia during the perioperative period attempts to maximize pain relief while minimizing adverse events from therapy. While various factors influence response to treatment among surgical patients, age, sex, race and pharmacogenetic differences appear to play major roles in predicting outcome. Genetic factors include a subset of genes that modulate the proteins involved in pain perception, pain pathway, analgesic metabolism (pharmacokinetics), transport and receptor signaling (pharmacodynamics). While results from adult genetic studies can provide direction for pediatric studies, they have limited direct applicability, as children's genetic predispositions to analgesic response may be influenced by developmental and behavioral components, altered sensitivity to analgesics and variation in gene-expression patterns. We have reviewed the available evidence on improving and personalizing pain management with opioids and the significance of individualizing analgesia, in order to maximize analgesic effect with minimal adverse effects with opioids. While the early evidence on individual genotype associations with pain, analgesia and opioid adverse outcome are promising, the large amount of conflicting data in the literature suggests that there is a need for larger and more robust studies with appropriate population stratification and consideration of nongenetic and other genetic risk factors. Although the clinical evidence and the prospect of being able to provide point-of-care genotyping to enable clinicians to deliver personalized analgesia for individual patients is still not available, positioning our research to identify all possible major genetic and nongenetic risk factors of an individual patient, advancing less expensive point-of-care genotyping technology and developing easy-to-use personalized clinical decision algorithms will help us to improve current clinical and economic outcomes associated with pain and opioid pain management.
Collapse
Affiliation(s)
- Senthilkumar Sadhasivam
- Department of Anesthesiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, MLC 2001, Cincinnati, OH 45229, USA.
| | | |
Collapse
|
66
|
Abstract
BACKGROUND The clinical utility of opioids is limited by adverse drug effects including respiratory depression, sedation, nausea, and pruritus. In addition, abuse of prescription opioids is problematic. Gaining a better understanding of the genetic and environmental mechanisms contributing to an individual's susceptibility to adverse opioid effects is essential to identify patients at risk. METHODS A classic twin study paradigm provided estimates for the genetic and familial (genetic and/or shared environment) contribution to acute adverse and affective opioid responses, all secondary outcomes of a larger dataset. One hundred twenty-one twin pairs were recruited in a single occasion, randomized, double-blind, and placebo-controlled study. The μ-opioid receptor agonist alfentanil and saline placebo were administered as target-controlled infusions under carefully monitored laboratory conditions. Measured outcomes included respiratory depression, sedation, nausea, pruritus, drug liking, and drug disliking. Demographic information was collected, and aspects of mood and sleep were evaluated. RESULTS Significant heritability was detected for respiratory depression (30%), nausea (59%), and drug disliking (36%). Significant familial effects were detected for sedation (29%), pruritus (38%), dizziness (32%), and drug liking (26%). Significant covariates included age, sex, race, ethnicity, education, mood, and depression. Covariates affected sedation, pruritus, drug liking and disliking, and dizziness. CONCLUSIONS This study demonstrates that large-scale efforts to collect quantitative and well-defined opioid response data are not only feasible but also produce data that are suitable for genetic analysis. Genetic, environmental, and demographic factors work together to control adverse and reinforcing opioid responses, but contribute differently to specific responses.
Collapse
|
67
|
De Gregori M, Garbin G, De Gregori S, Minella CE, Bugada D, Lisa A, Govoni S, Regazzi M, Allegri M, Ranzani GN. Genetic variability at COMT but not at OPRM1 and UGT2B7 loci modulates morphine analgesic response in acute postoperative pain. Eur J Clin Pharmacol 2013; 69:1651-8. [DOI: 10.1007/s00228-013-1523-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 04/24/2013] [Indexed: 10/26/2022]
|
68
|
Mura E, Govoni S, Racchi M, Carossa V, Ranzani GN, Allegri M, van Schaik RH. Consequences of the 118A>G polymorphism in the OPRM1 gene: translation from bench to bedside? J Pain Res 2013; 6:331-53. [PMID: 23658496 PMCID: PMC3645947 DOI: 10.2147/jpr.s42040] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The 118A>G single nucleotide polymorphism (SNP) in the μ-opioid receptor (OPRM1) gene has been the most described variant in pharmacogenetic studies regarding opioid drugs. Despite evidence for an altered biological function encoded by this variant, this knowledge is not yet utilized clinically. The aim of the present review was to collect and discuss the available information on the 118A>G SNP in the OPRM1 gene, at the molecular level and in its clinical manifestations. In vitro biochemical and molecular assays have shown that the variant receptor has higher binding affinity for β-endorphins, that it has altered signal transduction cascade, and that it has a lower expression compared with wild-type OPRM1. Studies using animal models for 118A>G have revealed a double effect of the variant receptor, with an apparent gain of function with respect to the response to endogenous opioids but a loss of function with exogenous administered opioid drugs. Although patients with this variant have shown a lower pain threshold and a higher drug consumption in order to achieve the analgesic effect, clinical experiences have demonstrated that patients carrying the variant allele are not affected by the increased opioid consumption in terms of side effects.
Collapse
Affiliation(s)
- Elisa Mura
- Department of Drug Sciences, Centre of Excellence in Applied Biology, University of Pavia, Pavia, Italy
| | | | | | | | | | | | | |
Collapse
|
69
|
Ginosar Y, Birnbach DJ, Shirov TT, Arheart K, Caraco Y, Davidson EM. Duration of analgesia and pruritus following intrathecal fentanyl for labour analgesia: no significant effect of A118G μ-opioid receptor polymorphism, but a marked effect of ethnically distinct hospital populations. Br J Anaesth 2013; 111:433-44. [PMID: 23592691 DOI: 10.1093/bja/aet075] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Genetic polymorphism (A118G) in the μ-opioid receptor has been reported to affect systemic opioid analgesia. However, reported pharmacogenetic effects on spinal opioid analgesia, particularly in labour, have been equivocal. METHODS We prospectively assessed effects of the μ-opioid receptor A118G single nucleotide polymorphism (SNP) on analgesia after 20 μg of spinal fentanyl. We studied two ethnically distinct hospital populations (Miami and Jerusalem). Independent variables were A118G, ethnicity, and hospital. Primary outcome was time from spinal analgesia until analgesic request. Secondary outcomes were pain and pruritus, assessed at repeated intervals until analgesia request. RESULTS One hundred and twenty-five nulliparous parturients in early labour were analysed. The allelic frequency of A118G was 14.8% (14.4% in Miami; 15.5% in Jerusalem). Time to analgesia request (sd) in Miami was 122 (44) min and in Jerusalem was 87 (32) min, P<0.001; Hispanic 123 (46) min vs Jew/Arab 87 (32) min, P<0.001; Black 121 (41) min vs Jew/Arab 87 (32) min, P=0.015. There was no significant effect of A118G. Survival analysis showed Miami > Jerusalem, P<0.001; Hispanics and Black > Jew/Arab, P<0.001; no effect of A118G. Within hospital groups, A118G had no effect on time to analgesic request; within genomic groups there was a significant difference between hospitals. The time-course for pruritus exactly paralleled the time-course for analgesia and was affected by hospital (P=0.006) and by ethnic group (P=0.03), but not by A118G. CONCLUSIONS We found no significant effect for the A118G single nucleotide polymorphism (SNP) on analgesic duration after spinal fentanyl for labour. In contrast, ethnically distinct hospital population groups exerted a marked effect on the time-course of both analgesia and pruritus.
Collapse
Affiliation(s)
- Y Ginosar
- Department of Anesthesiology and Critical Care Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel.
| | | | | | | | | | | |
Collapse
|
70
|
Kolesnikov Y, Gabovits B, Levin A, Veske A, Qin L, Dai F, Belfer I. Chronic pain after lower abdominal surgery: do catechol-O-methyl transferase/opioid receptor μ-1 polymorphisms contribute? Mol Pain 2013; 9:19. [PMID: 23566343 PMCID: PMC3623849 DOI: 10.1186/1744-8069-9-19] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 04/03/2013] [Indexed: 11/12/2022] Open
Abstract
Background Preoperative pain, type of operation and anesthesia, severity of acute postoperative pain, and psychosocial factors have been identified as risk factors for chronic postsurgical pain (CPP). Recently, it has been suggested that genetic factors also contribute to CPP. In this study, we aimed to determine whether the catechol-O-methyl transferase (COMT) and opioid receptor μ-1 (OPRM1) common functional polymorphisms rs4680 and rs1799971 were associated with the incidence, intensity, or duration of CPP in patients after lower abdominal surgery. Methods One hundred and two patients with American Society of Anesthesiologists (ASA) physical status I/II underwent either abdominal radical prostatectomy (n = 45) or hysterectomy (n = 57). The incidences of CPP in the pelvic and scar areas were evaluated in all patients three months after surgery. Results Thirty-five (34.3%) patients experienced CPP after lower abdominal surgery. Within this group, six (17.1%) patients demonstrated symptoms of neuropathic pain. For COMT rs4680, 22 (21.6%) patients had Met158Met, 55 (53.9%) patients had Val158Met, and 25 (24.5%) patients had Val158Val. No association was found between CPP phenotypes (incidence, intensity, and duration) and different rs4680 genotypes. For OPRM1 rs1799971, only CPP patients carrying at least one copy of the G allele had higher pain intensity than A118A carriers (p=0.02). No associations with other phenotypes were found. No combined effect of COMT/OPRM1 polymorphisms on CPP phenotypes was observed. Conclusions OPRM1 genotype influences CPP following lower abdominal surgery. COMT didn’t affect CPP, suggesting its potential modality-specific effects on human pain.
Collapse
Affiliation(s)
- Yuri Kolesnikov
- East Tallinn Central Hospital, Tallinn Technological University, Tallinn, Estonia
| | | | | | | | | | | | | |
Collapse
|
71
|
Belfer I. Nature and nurture of human pain. SCIENTIFICA 2013; 2013:415279. [PMID: 24278778 PMCID: PMC3820306 DOI: 10.1155/2013/415279] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Accepted: 03/14/2013] [Indexed: 05/05/2023]
Abstract
Humans are very different when it comes to pain. Some get painful piercings and tattoos; others can not stand even a flu shot. Interindividual variability is one of the main characteristics of human pain on every level including the processing of nociceptive impulses at the periphery, modification of pain signal in the central nervous system, perception of pain, and response to analgesic strategies. As for many other complex behaviors, the sources of this variability come from both nurture (environment) and nature (genes). Here, I will discuss how these factors contribute to human pain separately and via interplay and how epigenetic mechanisms add to the complexity of their effects.
Collapse
Affiliation(s)
- Inna Belfer
- Departments of Anesthesiology and Human Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
72
|
Hajj A, Khabbaz L, Laplanche JL, Peoc’h K. Pharmacogenetics of opiates in clinical practice: the visible tip of the iceberg. Pharmacogenomics 2013; 14:575-85. [DOI: 10.2217/pgs.13.13] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Opioids are the cornerstone of analgesic therapy and are used as a substitution therapy for opiate addiction. Interindividual variability in response to opioids is a significant challenge in the management of pain and substitution. Therefore, treatment with opioids requires a careful individualization of dosage to achieve an appropriate balance of efficacy and adverse effects and, consequently, avoid toxicity, particularly respiratory depression, sedation and for some, cardiac ventricular fibrillations. Many studies have investigated the association between genetic factors and the variability of response to opioids. Variants in genes encoding proteins implied in opioid pharmacokinetics (absorption, distribution, metabolism, excretion and toxicity), together with those implied in opioids direct and indirect pharmacodynamics (genes of opioid receptors and monoaminergic systems), are the most studied. Many association studies have not been replicated. The purpose of this article is to summarize pharmacogenetic data associated with some opioids frequently encountered in managed care settings.
Collapse
Affiliation(s)
- Aline Hajj
- Laboratoire de Pharmacologie Clinique et Pharmacocinétique, Faculté de Pharmacie, Université Saint Joseph, Beyrouth, Liban, Lebanon
| | - Lydia Khabbaz
- Laboratoire de Pharmacologie Clinique et Pharmacocinétique, Faculté de Pharmacie, Université Saint Joseph, Beyrouth, Liban, Lebanon
| | - Jean-Louis Laplanche
- Service de Biochimie et de Biologie moléculaire, Hôpital Lariboisière, AP-HP, Paris, France
- INSERMU705/UMR8206 & Laboratoire de Biologie Cellulaire, Université Paris Descartes, 6 Avenue de l’Observatoire, 75006 Paris, France
| | - Katell Peoc’h
- INSERMU705/UMR8206 & Laboratoire de Biologie Cellulaire, Université Paris Descartes, 6 Avenue de l’Observatoire, 75006 Paris, France
- Service de Biochimie et de Biologie moléculaire, Hôpital Lariboisière, AP-HP, Paris, France.
| |
Collapse
|
73
|
Catechol-O-methyltransferase gene polymorphism and chronic human pain: a systematic review and meta-analysis. Pharmacogenet Genomics 2013; 22:673-91. [PMID: 22722321 DOI: 10.1097/fpc.0b013e3283560c46] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
In human studies, low COMT (catechol-O-methyltransferase) activity has been associated with increased sensitivity to acute clinical preoperative or postoperative pain. We explored the association between the COMT genotype and three chronic pain conditions: migrainous headache, fibromyalgia, or chronic widespread pain and chronic musculoskeletal pain. Furthermore, we evaluated whether COMT genotype affects the efficacy of opioids in chronic pain. After a systematic literature review, we carried out meta-analyses on the three chronic pain conditions. The efficacy of opioids was evaluated using a systematic review only. The meta-analyses showed that fibromyalgia or chronic widespread pain is the only type of chronic pain that could be associated with the COMT single nucleotide polymorphism rs4680 (Val158Met). Met158, which results in the low-activity variant of COMT, is the risk allele. In chronic clinical pain, the effect of the COMT polymorphism depends on the pain condition. Low COMT activity is not associated with migrainous headache or chronic musculoskeletal pain conditions, but it may increase the risk for fibromyalgia or chronic widespread pain. Low COMT activity increases opioid receptors and enhances opioid analgesia and adverse effects in some cancer pains. Findings from animal studies that have utilized COMT inhibitors elucidate the mechanism behind these findings. In rodent pain models, COMT inhibitors are pronociceptive, except for neuropathic pain models, where nitecapone was found to be antiallodynic. The complex interplay between enhanced adrenergic and dopaminergic activity in different parts of the nociceptive system probably explains the complicated actions of low COMT activity.
Collapse
|
74
|
Dalal PG, Doheny KK, Klick L, Britcher S, Rebstock S, Bezinover D, Palmer C, Berlin C, Postula M, Kong L, Janicki PK. Analysis of acute pain scores and skin conductance measurements in infants. Early Hum Dev 2013; 89:153-8. [PMID: 23046994 DOI: 10.1016/j.earlhumdev.2012.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 09/11/2012] [Accepted: 09/14/2012] [Indexed: 12/01/2022]
Abstract
BACKGROUND Skin conductance (SC) has been previously used to measure acute post-operative pain in adults and older children (>1year old).We have investigated the ability of SC to predict the severity of post-operative pain scores in the exclusively infant population. METHODS Infants (ages 6-12months) scheduled for elective surgery were recruited for the study. Data for behavioral pain scores and SC values - frequency of electrodermal responses per second (EDR/s), peak and basal levels, were recorded in the post-anesthesia care unit (PACU). Blood samples were collected for genomic studies, including single nucleotide polymorphisms (SNP) in morphine opioid receptor (MOR) A118G and the catechol-O-methyltransferase (COMT) G1947A genes. RESULTS 31 infants, mean age 8.9months (±1.9); mean weight 8.5kg (±1.1) were included in the final study analysis. With every 0.1 unit increase in peak values noted on SC, the odds of higher pain scores were found to be 5% greater (p=0.03). For predictability of moderate to severe pain, the area under the curve, sensitivity and specificity were 0.64, 90.9% and 51.4% respectively for peak values and 0.66, 54.5% and 79.4% respectively for EDR/s values. Genotyping performed in 16 out of 31 infants demonstrated that the carriers of MOR 118G allele had consistently higher basal SC values in the PACU. CONCLUSION Peak SC values may serve as indicators of unmitigated pain. Further studies are needed to fully investigate the effect of MOR A118G SNP on the post operative pain scores and SC values in the larger infant population in order to validate both the clinical significance of the skin conductance for routine pain assessment in infants and the observed genetic effect.
Collapse
Affiliation(s)
- Priti G Dalal
- Department of Anesthesiology, Penn State Milton S Hershey Medical Center and Penn State College of Medicine, Hershey, PA 17033, United States.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
|
76
|
The Effect of OPRM1 and COMT Genotypes on the Analgesic Response to Intravenous Fentanyl Labor Analgesia. Anesth Analg 2013; 116:386-91. [DOI: 10.1213/ane.0b013e318273f2c7] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
77
|
Liao Q, Chen DJ, Zhang F, Li L, Hu R, Tang YZ, Ou-Yang W, Huang D. Effect of CYP3A4*18B polymorphisms and interactions with OPRM1 A118G on postoperative fentanyl requirements in patients undergoing radical gastrectomy. Mol Med Rep 2013; 7:901-8. [PMID: 23313934 DOI: 10.3892/mmr.2013.1270] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 11/26/2012] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to investigate the effect of cytochrome P450 3A4 (CYP3A4)*18B polymorphisms and the interaction of the µ opioid receptor gene (OPRM1) A118G and CYP3A4*18B polymorphisms on postoperative fentanyl analgesia in Chinese Han patients undergoing radical gastrectomy. In total, 97 patients scheduled to undergo radical gastrectomy under general anesthesia were enrolled in this study. Post‑operative patient‑controlled intravenous analgesia of fentanyl was administered as analgesia up to 48 h following surgery. Venous blood (2 ml) was obtained from each patient to measure the OPRM1 A118G and CYP3A4*18B genotypes. The differences in fentanyl consumption and adverse effects were compared among the genotypes at 24 and 48 h following surgery. In the first 48 h following surgery, patients in the CYP3A4*18B/*18B group consumed significantly less fentanyl compared with patients in the *1/*1 group (P=0.032). With regards to the joint genetic effect, during the 48‑h period, patients with AA and *1*18B polymorphisms received fewer fentanyl doses compared with those with AG and *1*1 (P=0.049), while patients with AG and *1*18B polymorphisms received significantly fewer fentanyl doses compared with those with AG and *1*1 (P=0.010), and patients with *18B*18B polymorphisms received significantly fewer fentanyl doses compared with those with AA and *1*1 (P=0.024) or those with AG and *1*1 polymorphisms (P=0.006). No correlation between OPRM1 A118G and CYP3A4*18B and postoperative nausea, vomiting and dizziness was found. Results demonstrated that 48 h following surgery, patients with the CYP3A4*18B/*18B genotype required less fentanyl than patients with the CYP3A4*1/*1 genotype to control pain. Additionally, the combined genotype of CYP3A4*18B and OPRM1 A118G may affect fentanyl doses administered for pain control, but not postoperative nausea, vomiting and dizziness.
Collapse
Affiliation(s)
- Qin Liao
- Department of Anesthesiology, The Third Xiangya Hospital of Central South University, Changsha 410013, PR China
| | | | | | | | | | | | | | | |
Collapse
|
78
|
Mamie C, Rebsamen MC, Morris MA, Morabia A. First Evidence of a Polygenic Susceptibility to Pain in a Pediatric Cohort. Anesth Analg 2013; 116:170-7. [DOI: 10.1213/ane.0b013e31826f0637] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
79
|
Beyaz SG. Comparison of efficacy of intra-articular morphine and steroid in patients with knee osteoarthritis. J Anaesthesiol Clin Pharmacol 2012; 28:496-500. [PMID: 23225932 PMCID: PMC3511949 DOI: 10.4103/0970-9185.101940] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Introduction: Primary therapeutic aim in treatment of osteoarthritis of the knee is to relieve the pain of osteoarthritis. The aim of this study was to compare the efficacy of intra-articular triamcinolone with intra-articular morphine in pain relief due to osteoarthritis of the knee in the elderly population. Materials and Methods: Patients between 50 and 80 years of age were randomized into three groups. Group M received morphine plus bupivacaine intra-articularly, Group T received triamcinolone plus bupivacaine intra-articularly, and Group C received saline plus bupivacaine intra-articularly. Patients were evaluated before injection and in 2nd, 4th, 6th, and 12th weeks after injection. First-line supplementary analgesic was oral paracetamol 1500 mg/day. If analgesia was insufficient with paracetamol, oral dexketoprofen trometamol 50 mg/day was recommended to patients. Results: After the intra-articular injection, there was statistically significant decrease in visual analog scale (VAS) scores in Groups M and T, when compared to Group C. The decrease of VAS scores seen at the first 2 weeks continued steadily up to the end of 12th week. There was a significant decrease in Groups M and T in the WOMAC scores, when compared to Group C. There was no significant difference in the WOMAC scores between morphine and steroid groups. Significantly less supplementary analgesics was used in the morphine and steroid groups. Conclusion: Intra-articular morphine was as effective as intra-articular triamcinolone for analgesia in patients with osteoarthritis knee. Intra-articular morphine is possibly a better option than intra-articular steroid as it has lesser side effects.
Collapse
Affiliation(s)
- Serbülent Gökhan Beyaz
- Department of Anesthesia and Pain Management Center, Sakarya University Medical School, Sakarya, Turkey
| |
Collapse
|
80
|
Abstract
This paper is the thirty-fourth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2011 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
81
|
Pang GSY, Ithnin F, Wong YY, Wang JB, Lim Y, Sia ATH, Lee CGL. A non-synonymous single nucleotide polymorphism in an OPRM1 splice variant is associated with fentanyl-induced emesis in women undergoing minor gynaecological surgery. PLoS One 2012; 7:e48416. [PMID: 23144877 PMCID: PMC3492352 DOI: 10.1371/journal.pone.0048416] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 09/25/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Fentanyl-induced emesis (FIE) is a distressing adverse effect in the postoperative setting. The genetic basis of FIE remains largely unknown, therefore, we examined whether it was associated with specific genetic variants of OPRM1, the gene encoding the main receptor target of fentanyl. METHODS In this prospective case-control study, 193 women undergoing gynaecological surgery under a standardized anaesthetic with a low emetogenic risk were enrolled. Inclusion and exclusion criteria were designed to select extreme phenotypes as well as to ensure that most major confounders for FIE were either excluded or present in all patients. To control for unforeseen intra- and postoperative confounders for FIE, only 161 patients were further analysed, out of which 10 were categorized as having FIE, defined by the presence of at least one of three symptoms: nausea, vomiting or retching that was likely to be fentanyl-related. To identify SNPs relevant to FIE in our population, DNA from 40 controls and 10 cases was sequenced at the following OPRM1 regions: 3 kbp of the promoter, main and alternative exons as well as 2 kbp of the 3' downstream region. The genotype of the significant SNP was further determined in the remaining 111 controls. RESULTS The incidence of FIE was 6.2%. Initial sequencing of 10 cases and 40 controls identified 25 SNPs. Only rs540825, a non-synonymous SNP in the splice variant, MOR1X, showed a significant association with FIE post-Bonferroni correction. This SNP was further examined in the remaining 111 controls which confirmed its significant association with FIE (p = 0.019 post-Bonferroni, OR: 5.6, 95% CI: 1.42-21.91). CONCLUSIONS This is the first report of an association between the occurrence of FIE in Chinese women undergoing gynaecological surgery and an OPRM1 splice variant SNP, rs540825.
Collapse
Affiliation(s)
- Grace Su Yin Pang
- Liver Cancer Functional Genomics Laboratory, National Cancer Centre Singapore, Singapore, Singapore
- Department of Palliative Medicine, National Cancer Centre Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Farida Ithnin
- Department of Obstetric Anaesthesia, KK Women's and Children's Hospital, Singapore, Singapore
| | - Yin Yee Wong
- Department of Palliative Medicine, National Cancer Centre Singapore, Singapore, Singapore
| | - Jing Bo Wang
- Liver Cancer Functional Genomics Laboratory, National Cancer Centre Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yvonne Lim
- Department of Obstetric Anaesthesia, KK Women's and Children's Hospital, Singapore, Singapore
| | - Alex Tiong Heng Sia
- Department of Obstetric Anaesthesia, KK Women's and Children's Hospital, Singapore, Singapore
| | - Caroline Guat Lay Lee
- Liver Cancer Functional Genomics Laboratory, National Cancer Centre Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- DUKE-NUS Graduate Medical School, Singapore, Singapore
| |
Collapse
|
82
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to highlight postoperative nausea and vomiting (PONV), to discuss why it occurs, how it might be prevented and then how it can be treated. RECENT FINDINGS PONV/postdischarge nausea and vomiting (PDNV) is a problem that patients identify as one of the worst, if not the worst problem that can occur after surgery. Therapy is not perfect, yet there are anaesthesia techniques that can help minimize the problem and drugs that can be used both to prevent and also treat the problem once it occurs. There is a genetic basis for why some people experience PONV/PDNV more than others and also why treatment for some is better than others. SUMMARY It is easy to turn the vapourizer dial, but that is a part of the problem. Not everyone reacts the same to drugs. Although PDNV is not as well characterized as PONV, at the very least, avoid an anaesthetic that may make PONV/PDNV worse and be aggressive in treating the problem.
Collapse
|
83
|
Vuilleumier PH, Stamer UM, Landau R. Pharmacogenomic considerations in opioid analgesia. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2012; 5:73-87. [PMID: 23226064 PMCID: PMC3513230 DOI: 10.2147/pgpm.s23422] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Indexed: 12/25/2022]
Abstract
Translating pharmacogenetics to clinical practice has been particularly challenging in the context of pain, due to the complexity of this multifaceted phenotype and the overall subjective nature of pain perception and response to analgesia. Overall, numerous genes involved with the pharmacokinetics and dynamics of opioids response are candidate genes in the context of opioid analgesia. The clinical relevance of CYP2D6 genotyping to predict analgesic outcomes is still relatively unknown; the two extremes in CYP2D6 genotype (ultrarapid and poor metabolism) seem to predict pain response and/or adverse effects. Overall, the level of evidence linking genetic variability (CYP2D6 and CYP3A4) to oxycodone response and phenotype (altered biotransformation of oxycodone into oxymorphone and overall clearance of oxycodone and oxymorphone) is strong; however, there has been no randomized clinical trial on the benefits of genetic testing prior to oxycodone therapy. On the other hand, predicting the analgesic response to morphine based on pharmacogenetic testing is more complex; though there was hope that simple genetic testing would allow tailoring morphine doses to provide optimal analgesia, this is unlikely to occur. A variety of polymorphisms clearly influence pain perception and behavior in response to pain. However, the response to analgesics also differs depending on the pain modality and the potential for repeated noxious stimuli, the opioid prescribed, and even its route of administration.
Collapse
Affiliation(s)
- Pascal H Vuilleumier
- Klinik für Anästhesiologie und Schmerztherapie, Inselspital Universität Bern, Switzerland
| | | | | |
Collapse
|
84
|
|
85
|
Branford R, Droney J, Ross JR. Opioid genetics: the key to personalized pain control? Clin Genet 2012; 82:301-10. [PMID: 22780883 DOI: 10.1111/j.1399-0004.2012.01923.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 06/25/2012] [Accepted: 06/29/2012] [Indexed: 11/28/2022]
Abstract
There are now several strong opioids available to choose from for the relief of moderate to severe pain. On a population level, there is no difference in terms of analgesic efficacy or adverse reactions between these drugs; however, on an individual level there is marked variation in response to a given opioid. The genetic influences to this variation are complex, and although current research has shown some promising results, these have not been replicated across larger studies and as such the ultimate aim of personalized prescribing remains elusive. If personalized prescribing could be achieved this would have a major impact at an individual level to facilitate safe, effective and rapid symptom control. This review presents some of the recent positive advances in opioid pharmacogenetic studies, focusing on associations between candidate genes and the three main elements of opioid response: analgesic, upper gastrointestinal and central adverse reactions.
Collapse
Affiliation(s)
- R Branford
- Department of Medicine, Royal Marsden NHS Foundation Trust, London, UK
| | | | | |
Collapse
|
86
|
Hastie BA, Riley JL, Kaplan L, Herrera DG, Campbell CM, Virtusio K, Mogil JS, Wallace MR, Fillingim RB. Ethnicity interacts with the OPRM1 gene in experimental pain sensitivity. Pain 2012; 153:1610-1619. [PMID: 22717102 DOI: 10.1016/j.pain.2012.03.022] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2011] [Revised: 02/28/2012] [Accepted: 03/21/2012] [Indexed: 01/13/2023]
Abstract
Robust interindividual variation in pain sensitivity has been observed, and recent evidence suggests that some of the variability may be genetically mediated. Our previous data revealed significantly higher pressure pain thresholds among individuals possessing the minor G allele of the A118G SNP of the mu-opioid receptor gene (OPRM1) compared with those with 2 consensus alleles. Moreover, ethnic differences in pain sensitivity have been widely reported. Yet, little is known about the potential interactive associations of ethnicity and genotype with pain perception. This study aimed to identify ethnic differences in OPRM1 allelic associations with experimental pain responses. A total of 247 healthy young adults from three ethnic groups (81 African Americans; 79 non-white Hispanics; and 87 non-Hispanic whites) underwent multiple experimental pain modalities (thermal, pressure, ischemic, cold pressor). Few African Americans (7.4%) expressed the rare allele of OPRM1 compared to non-Hispanic whites and Hispanics (28.7% vs. 27.8%, respectively). Across the entire sample, OPRM1 genotype did not significantly affect pain sensitivity. However, analysis in each ethnic group separately revealed significant genotype effects for most pain modalities among non-Hispanic-whites (P<.05) but not Hispanics or African Americans. The G allele was associated with decreased pain sensitivity among whites only; a trend in the opposite direction emerged in Hispanics. The reasons for this dichotomy are unclear; they may involve ethnic differences in haplotypic structure, or A118G may be a tag-SNP linked to other functional polymorphisms. These findings demonstrate an ethnicity-dependent association of OPRM1 genotype with pain sensitivity. Additional research is warranted to uncover the mechanisms influencing these relationships.
Collapse
Affiliation(s)
- Barbara A Hastie
- Department of Community Dentistry and Behavioral Science, University of Florida College of Dentistry, Gainesville, FL, USA Genoptix Medical Laboratory, Carlsbad, CA, USA Centers for Disease Control and Prevention, Atlanta, GA, USA Department of Psychiatry and Behavioral Science, Johns Hopkins University, Baltimore, MD, USA Department of Psychology and Alan Edwards Centre for Research on Pain, McGill University, Montreal, Quebec, Canada Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL, USA Malcom Randall V.A. Medical Center, Gainesville, FL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Abstract
PURPOSE OF REVIEW On a population level, there is no difference in terms of efficacy or side-effects between any of the strong opioids. On an individual level, however, there is marked variation in response to opioids. This review presents some of the recent advances in opioid pharmacogenetic studies. RECENT FINDINGS A growing number of genes have been studied in a number of different patient populations. Most data have come from candidate-gene studies. There have been two genome-wide association studies in pain and opioid response. The clinical and genetic complexity of response to opioids has limited the clinical applicability of the genetic results. Currently, interindividual variation in opioid response is managed clinically through a process known as opioid switching. The evidence supporting the efficacy of opioid switching is poor, mainly because randomized controlled trials in this area are lacking. SUMMARY Adequately powered studies to allow identification of genetic variants with small effect size and exploration of gene-gene interaction are needed. Integration of genetic analysis in clinical studies with carefully defined outcome measures will increase the likelihood of identifying clinical and genetic factors which can be used to predict opioid response.
Collapse
|
88
|
Mogil JS. Pain genetics: past, present and future. Trends Genet 2012; 28:258-66. [PMID: 22464640 DOI: 10.1016/j.tig.2012.02.004] [Citation(s) in RCA: 226] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 02/16/2012] [Accepted: 02/22/2012] [Indexed: 01/01/2023]
Abstract
Chronic pain is a classic example of gene × environment interaction: inflammatory and/or nerve injuries are known or suspected to be the etiology of most chronic pain syndromes, but only a small minority of those subjected to such injuries actually develop chronic pain. Once chronic pain has developed, pain severity and analgesic response are also highly variable among individuals. Although animal genetics studies have been ongoing for over two decades, only recently have comprehensive human twin studies and large-scale association studies been performed. Here, I review recent and accelerating progress in, and continuing challenges to, the identification of genes contributing to such variability. Success in this endeavor will hopefully lead to both better management of pain using currently available therapies and the development and/or prioritizing of new ones.
Collapse
Affiliation(s)
- Jeffrey S Mogil
- Department of Psychology, McGill University, Montreal, QC, H3A 1B1, Canada.
| |
Collapse
|
89
|
Pharmacological consequence of the A118G μ opioid receptor polymorphism on morphine- and fentanyl-mediated modulation of Ca²⁺ channels in humanized mouse sensory neurons. Anesthesiology 2011; 115:1054-62. [PMID: 21926562 DOI: 10.1097/aln.0b013e318231fc11] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
BACKGROUND The most common functional single nucleotide polymorphism of the human OPRM1 gene, A118G, has been shown to be associated with interindividual differences in opioid analgesic requirements, particularly with morphine, in patients with acute postoperative pain. The purpose of this study was to examine whether this polymorphism would modulate the morphine and fentanyl pharmacological profile of sensory neurons isolated from a humanized mouse model homozygous for either the 118A or 118G allele. METHODS The coupling of wild-type and mutant μ opioid receptors to voltage-gated Ca channels after exposure to either ligand was examined by employing the whole cell variant of the patch-clamp technique in acutely dissociated trigeminal ganglion neurons. Morphine-mediated antinociception was measured in mice carrying either the 118AA or 118GG allele. RESULTS The biophysical parameters (cell size, current density, and peak current amplitude potential) measured from both groups of sensory neurons were not significantly different. In 118GG neurons, morphine was approximately fivefold less potent and 26% less efficacious than that observed in 118AA neurons. On the other hand, the potency and efficacy of fentanyl were similar for both groups of neurons. Morphine-mediated analgesia in 118GG mice was significantly reduced compared with the 118AA mice. CONCLUSIONS This study provides evidence to suggest that the diminished clinical effect observed with morphine in 118G carriers results from an alteration of the receptor's pharmacology in sensory neurons. In addition, the impaired analgesic response with morphine may explain why carriers of this receptor variant have an increased susceptibility to become addicted to opioids.
Collapse
|
90
|
Abstract
An estimated 15-50% of the population experiences pain at any given time, at great personal and societal cost. Pain is the most common reason patients seek medical attention, and there is a high degree of individual variability in reporting the incidence and severity of symptoms. Research suggests that pain sensitivity and risk for chronic pain are complex heritable traits of polygenic origin. Animal studies and candidate gene testing in humans have provided some progress in understanding the heritability of pain, but the application of the genome-wide association methodology offers a new tool for further elucidating the genetic contributions to normal pain responding and pain in clinical populations. Although the determination of the genetics of pain is still in its infancy, it is clear that a number of genes play a critical role in determining pain sensitivity or susceptibility to chronic pain. This review presents an update of the most recent findings that associate genetic variation with variability in pain and an overview of the candidate genes with the highest translational potential.
Collapse
Affiliation(s)
- Erin E Young
- Department of Anesthesiology, Molecular Epidemiology of Pain Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | |
Collapse
|
91
|
Landau R, Ortner C, Carvalho B. Challenges in Interpreting Joined Allelic Combinations of OPRM1 and COMT Genes. Anesth Analg 2011; 113:432. [DOI: 10.1213/ane.0b013e31821f154f] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
92
|
Wildgaard K, Kehlet H. Chronic post-thoracotomy pain—What is new in pathogenic mechanisms and strategies for prevention? ACTA ACUST UNITED AC 2011. [DOI: 10.1053/j.trap.2011.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|