Prospective replication study implicates the catechol-O-methyltransferase Val158Met polymorphism as a biomarker for the response to morphine in patients with cancer

  • Authors:
    • Hiromichi Matsuoka
    • Chihiro Makimura
    • Atsuko Koyama
    • Yoshihiko Fujita
    • Junji Tsurutani
    • Kiyohiro Sakai
    • Ryo Sakamoto
    • Kazuto Nishio
    • Kazuhiko Nakagawa
  • View Affiliations

  • Published online on: August 8, 2017     https://doi.org/10.3892/br.2017.963
  • Pages: 380-384
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Genetic differences in humans cause clinical difficulties in opioid treatment. Previous studies indicate that a single nucleotide polymorphism in the catechol‑O‑methyltransferase (COMT) gene (rs4680; p.Val158Met) may present as a predictive biomarker for the response to morphine treatment. In our previous pilot exploratory study, patients with a G/G genotype were demonstrated to require a higher dose of morphine, compared with patients with A/A and A/G genotypes. In the present study, the aim was to replicate the findings in an independent cohort of opioid‑treatment‑naïve patients exhibiting various types of cancer. This prospective study was conducted from 2011 to 2012 at the Kindai University Faculty of Medicine. A total of 50 patients with opioid‑treatment naïve and histologically confirmed malignant neoplasms who were scheduled to undergo opioid treatment were evaluated in the present study. Assessments were conducted pre‑treatment (day 1), post‑treatment (day 1), and one week after treatment (day 8). The required dose of morphine on day 1 was significantly higher for patients with the G/G genotype of COMT, compared with those with the A/A and A/G genotypes (P=0.013). The results of the present study provide additional evidence that the COMT genotype may be a predictive biomarker for the response to morphine treatment.

Introduction

Pain is a feared and common complication in patients with cancer (1,2), which increases in prevalence and intensity with disease progression (3,4), and influences multiple aspects of quality of life (58). Pharmacogenetic, pharmacokinetic and pharmacodynamic variations among individuals result in a wide variety of responses to pain sensation and analgesics; therefore, investigations of biomarkers for opioid treatment have been performed to improve the efficacy of morphine treatment (9). According to a review published in 2015 (10), numerous studies have proposed that the catechol-O-methyltransferase (COMT) 472G→A (rs4680, p.Val158Met) genotype may be a predictive biomarker for the response to morphine treatment. In these studies, patients with the G/G genotype received the highest dose of morphine (11,12), while those with the A/A genotype received the lowest morphine dose (13). However, the majority of these studies had a small sample size, were retrospective, or were targeting non-cancer pain (14). A large sample study has indicated that none of the 112 single nucleotide polymorphisms (SNPs) in 25 candidate genes demonstrated significant associations with opioid dose (15); however, the quantity of concomitant analgesics [non-steroidal anti-inflammatory drugs (NSAIDs) and acetaminophen] and the method for dose determination of opioids remain unclear. Furthermore, the mean Brief Pain Index (16) following treatment was relatively high (15).

An exploratory study was planned, which included 100 patients with opioid-treatment-naïve cancer, in whom the use of NSAIDs and acetaminophen, as well as the method of dose determination of morphine were kept constant. The investigation was divided into a pilot study of 50 cases, and a replication study (the current study) of 50 more cases. Expression levels of functional genetic variants as predictive biomarkers of the response to morphine were examined. The pilot study indicated that COMT 472G→A may be a predictive biomarker (17). The aim of the current prospective study was to replicate these findings in an independent cohort of 50 opioid-treatment-naïve patients exhibiting various types of cancer.

Materials and methods

Patients and samples

This prospective study was conducted from April 2011 to March 2012 at the Kindai University Faculty of Medicine (Osaka, Japan). A total of 50 patients with opioid-treatment naïve and histologically confirmed malignant neoplasms who were scheduled to undergo opioid treatment were evaluated in the study. These patients were recruited and selected by non-probabilistic convenience sampling by their physician from the outpatients and inpatients service of the Department of Medical Oncology (Kindai University Faculty of Medicine). All patients met the following inclusion criteria: Aged ≥18 years, a verified cancer diagnosis, cancer pain suitable for morphine treatment (excluding patients with neuropathic pain, predominant incidental pain, glomerular filtration rate <30 ml/min, and history of opioid/drug abuse or alcoholism from a self-reported questionnaire), daily use of NSAIDs and able to provide written informed consent. The required dose of morphine (day 1) was evaluated in 50 patients, and was assessed in 48 patients, (2 were excluded who could not continuously receive morphine due to adverse effects or mortality; Fig. 1).

Clinical features, including age, sex, Eastern Cooperative Oncology Group performance status (PS) (18), and types of primary malignant neoplasm were recorded. Morphine was administered using the standard method, including titration [NCCN Guidelines (19), Adult Cancer Pain], performed by specialized palliative care doctors. Immediate release (IR) morphine (5 mg) was administered orally, and efficacy and adverse effects were reassessed at 60 min. When the pain score was not decreased, the same dose of IR morphine (5 mg) was re-administered. Dose titration was performed to decrease pain by ≥33% on the numerical rating scale (NRS) (20) pain scale at day 1 (pre-treatment), as well as to reduce the NRS to a score of ≤3.

Criteria for discontinuation of dose titration were appearance of adverse effects graded ≥3 (Common Terminology Criteria for Adverse Events, v4.0) (21), or an attending physician determining titration to be difficult to continue, even if the adverse effect was graded ≤2. Controlled release (CR) morphine, six times the dose of IR morphine, was administered orally once per day. If pain was insufficiently controlled by 10 mg IR morphine in total, 60 mg CR morphine was administered at night on day 1. The attending physician could reduce the dose of CR morphine based on the general condition of the patient. Morphine administration was discontinued following a side effect of grade ≥3. Standardized information on potential benefits and adverse effects was provided to the patients. The study was approved by the Regional Committee for Medical Research Ethics, Kindai University Faculty of Medicine. Informed consent was obtained from all participants in the study. All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional research committee and the 1964 Helsinki declaration and its later amendments or comparable ethical standards.

Pain score

The pain score was assessed using a 0–10 NRS (0, no pain to 10, worst imaginable pain) on day 1 (pre-treatment), day 1 (post-treatment) and day 8 (one week after treatment) by asking patients the following question: “How intense was your average pain for the past 24 h?”

Genotyping

SNPs of COMT 472G→A (rs4680; p.Val158Met) modulate the genetic response to opioid medications (22). In an exploratory study of 50 cases, a functional genotype analysis of opioid receptor µ 1 118A→G (rs1799971; p.Asn40Asp) and COMT 472G→A (rs4680; p.Val158Met) was performed to establish the predictive biomarkers for the treatment outcome of morphine, based on the plasma level of morphine and the required dose according to genotype on days 1, 2 and 8. On day 1, blood was sampled for genotyping, and the pain score was recorded on days 1 (before and after treatment), 2 and 8. The required dose of morphine on day 1 was significantly higher in patients with the COMT G/G genotype, compared with those with A/A and A/G genotypes in the previous pilot study (P=0.03) (16). Therefore, in the current prospective study of 50 independent cases, COMT alone was the main focus.

Genomic DNA was isolated from blood samples using a QIAamp(r) DNA Mini kit (cat. no. 51304; Qiagen GmbH, Hilden, Germany) and was subjected to a genotype analysis using Taqman® SNP Genotyping Assay for COMT rs4680 (cat. no.4362691; Thermo Fisher Scientific Inc., Waltham, MA, USA) and a StepOnePlus™ Real-Time PCR Systems (cat no. 376600; Thermo Fisher Scientific Inc.).

Statistical analysis

All data are expressed as means ± standard deviation. Differences in the effects of morphine between gene polymorphism groups were evaluated using a Mann-Whitney U test. Two-sided P<0.05 was considered to indicate a statistically significant difference. All analyses were performed using SPSS software (v19.0; IBM SPSS, Armonk, NY, USA).

Results

Patient characteristics

The characteristics of the 50 patients (25 men and 25 women) are presented in Table I. The median age was 63 years (age, 36–80 years; 62±9.17). The primary tumors were as follows: Lung cancer, n=19 (38%); breast cancer, n=9 (18%); colorectal cancer, n=5 (10%); head and neck cancer, n=4 (8%); gastric cancer, n=3 (6%); unknown primary cause, n=3 (6%); pancreatic cancer, n=2 (4%); gallbladder cancer, n=2 (4%); and others, n=3 (6%). More than 80% of the subjects exhibited metastasis and a PS of 0 to 2. The median required doses of morphine were 31.2 mg (20–60 mg) on day 1 and 32.6 mg (20–60 mg) on day 8. The pain NRS before and after treatment did not differ between the G/G and non-G/G groups (P=0.177). A dose of 60 mg morphine was required in 4 cases of lung cancer and 1 case of unknown primary cancer. The causes of pain were lymph node metastases and bone metastases in 4 cases each, and pleural seeding in 1 case. Features of 5 high-dose morphine-requiring cases, defined as requiring 2 or more cycles of 5 mg IR morphine, are presented in Table II.

Table I.

Clinical characteristics of the patients (n=50).

Table I.

Clinical characteristics of the patients (n=50).

CharacteristicsPatients (n)
Age, years
  <6527
  ≥6523
Sex
  Male25
  Female25
Performance status
  0–240
  3–410
Genotype: COMT 472G→A (rs4680; p.Val158Met)
  G/G29
  A/G19
  A/A2
Tumor type
  Lung19
  Breast9
  Colorectal5
  Head and neck4
  Gastric3
  Unknown primary3
  Gallbladder2
  Pancreas2
  Others3
Required dose of morphine on day 1 (mg)
  202
  3043
  605
Required dose of morphine on day 8 (mg)
  As required8
  203
  3030
  606
  901
  N.E.2
Day 1 (pre-treatment) pain NRS, mean (SD)
  All patients6.88 (2.43)
  G/G7.07 (2.25)
  Non-G/G (A/A and A/G)6.62 (2.69)
Day 1 (post-treatment) pain NRS, mean (SD)
  All patients2.46 (1.46)
  G/G2.38 (1.45)
  Non-G/G (A/A and A/G)2.57 (1.50)
Day 8 pain NRS, mean (SD)
  All patients3.60 (2.73)
  G/G3.17 (2.63)
  Non-G/G (A/A and A/G)4.19 (2.80)

[i] COMT, catechol-O-methyltransferase; N.E, not evaluated; NRS, numerical rating scale; SD, standard deviation.

Table II.

Characteristics of patients with high-dose morphine-requiring cases.

Table II.

Characteristics of patients with high-dose morphine-requiring cases.

PatientAge (y)SexPSCOMT genotypeTumor typePain NRS (before administration)
163M1G/GLung8
259M1G/GLung7
370M2G/GLung10
464F1G/GUnknown primary8
560F2G/GLung8

[i] PS, performance status; COMT, catechol-O-methyltransferase; NRS, numerical rating scale.

Pain titration

Morphine at the required dose was administered to patients as a controlled-release agent. On day 1, dose titration was performed to reach NRS ≤3 and pain control ≥33%. Titration was successful in 76% of all cases, but unsuccessful in 60% of G/G genotype cases.

Genotypes of COMT and treatment outcomes of morphine

A functional genotype analysis of COMT 472G→A (rs4680; p.Val158Met) was performed to establish the predictive biomarkers for the outcome of morphine treatment (Fig. 2). The outcome based on the required morphine dose on days 1 and 8 was examined according to the genotype. The required dose of morphine (mg) on day 1 in GG, AG, and AA were 35.2±11.5, 29.5±2.3, and 25.0±7.1, respectively, and was significantly higher for patients with the COMT G/G genotype, compared with those with non-G/G (A/A and A/G) genotypes (P=0.013).

Discussion

To the best of our knowledge, the present study is the first to demonstrate the reproducibility of differences between required doses of morphine among individuals, due to genetic polymorphisms, in an independent cohort of opioid-treatment-naïve patients exhibiting various types of cancer. The most important finding is that patients with the COMT G/G genotype require a higher dose of morphine (day 1), compared with patients with A/A and A/G genotypes.

The morphine effect varies due to genetic differences; therefore, genetic polymorphisms are considered to be predictive markers. Various clinical studies have demonstrated that the required dose of morphine is higher in subjects with a COMT G/G genotype, compared with other genotypes, in cancer and non-cancer pain (1114). The current findings are consistent with these results. A large sample study has indicated that none of 112 SNPs in the 25 candidate genes showed significant associations with opioid dose (15); however, this study lacked clarity regarding the doses of concomitant analgesics, the dose determination of opioids, and the high average total dose of morphine of 90 mg/day. Despite this dose, residual pain was relatively high (pain NRS in the past 24 h, 3.20±2.10 subsequent to analgesic use and no data for pain NRS prior to analgesic use) (15). This suggests a high proportion of refractory pain. By contrast, in the current study, the dose of concomitant analgesics was controlled (all patients received daily regular doses of NSAIDs and acetaminophen), the method of dose titration was consistent, and pain was relatively well-controlled (pain NRS following treatment, 2.46±1.46). Patients who report a higher NRS for baseline pain generally exhibit a larger change in raw pain intensity (23). The patients in the present study had relatively strong pain (pain NRS in the past 24 h, 6.88±2.43) that was relatively well-controlled, and this may have made it easier to identify a difference due to genetic polymorphism.

Notably, the required dose of morphine on day 1 was significantly higher in patients with the COMT G/G genotype. The enzyme activity of COMT is defined as high in G/G, intermediate in G/A and low in A/A. Impairment of the COMT enzyme suppresses the production of enkephalin, which causes subsequent opioid receptor expression upregulation (10). In the A/A genotype, the function of the µ receptor is weak and patients tend to be sensitive to pain. However, the density of µ receptors is proposed to be increased in this genotype, which may be the reason for the low quantity of morphine required (24,25).

The nature of the pain NRS questionnaire may also be relevant. As patients with the COMT A/A genotype are more sensitive to pain, the reported NRS may be higher than that for the quantity of morphine required (26). A meta-analysis by Lee et al (27) revealed an association between fibromyalgia and the COMT A/A genotype.

The present study has various limitations. First, it is not possible to rule out that differences in patients’ background between the COMT G/G genotype and COMT non-G/G genotype groups may be responsible for the higher required dose of morphine in the G/G group. Although there was no significant difference in age between the G/G genotype and non-G/G genotype groups (P=0.658), the PS tended to be poor in the G/G genotype group (P=0.053). However, there were no patients with poor PS (3 and 4) in the high-dose morphine group. The COMT G/G genotype group has a marginally higher NRS score (prior to treatment) than the non-G/G genotype group. Although there was no significant difference in the NRS score before treatment between these groups (P=0.177), NRS scores tended to be higher in high-dose morphine-requiring cases compared with other cases. Thus, patient backgrounds require evaluation in future studies. In addition, the patients were included in this study based on the decision of attending physicians in a single hospital; consequently, the results may not be generalizable to other centers. Furthermore, disease duration, comorbidity, use of drugs other than opioids, NSAIDs, acetaminophen, and psychosocial factors, including education level and occupation, were not examined. Finally, the sample size is not considered to be large enough for a genetic study, which increases the rate of false positives. Therefore, the current results should be treated with caution until the present study is repeated using a larger population sample.

In conclusion, within these limitations, these results indicate that the COMT genotype influences the outcome of morphine treatment; therefore, it may be useful as a predictive biomarker for morphine treatment. The frequency of the GG allele has been found to be around 20% in previous studies (11,12,25), but close to 50% in a study targeting Japanese patients (17). This demonstrates ethnic differences in allele frequencies. This observation supports that it is clinically meaningful not to choose morphine as the first opioid in G/G patients with cancer pain, as higher doses may be required. However, these findings are from a prospective observational study, and an interventional study is required for further evaluation.

Acknowledgements

The present study was supported by the Third-Term Comprehensive 10-Year Strategy for Cancer Control and a Grant-in-Aid for Cancer Research from the Ministry of Health, Labour and Welfare (grant no. H22-037). The authors would like to thank Mrs. Tomoko Kitayama, Mrs. Erina Hatashita, Mrs. Kiyoko Kuwata, Mrs. Haruka Yamaguchi, Mr. Hiromasa Wadaguri and Mrs. Akiko Mizumoto for their technical assistance.

References

1 

Cleeland CS: Undertreatment of cancer pain in elderly patients. JAMA. 279:1914–1915. 1998. View Article : Google Scholar : PubMed/NCBI

2 

Patrick DL, Ferketich SL, Frame PS, Harris JJ, Hendricks CB, Levin B, Link MP, Lustig C, McLaughlin J, Reid LD, et al: National Institutes of Health State-of-the-Science Panel: National Institutes of Health State-of-the-Science Conference Statement: Symptom management in cancer: pain, depression, and fatigue. July;15–17. 2002.J Natl Cancer Inst Monogr. 32:9–16, 2004.

3 

Potter J and Higginson IJ: Pain experienced by lung cancer patients: A review of prevalence, causes and pathophysiology. Lung Cancer. 43:247–257. 2004. View Article : Google Scholar : PubMed/NCBI

4 

Butler LD, Koopman C, Cordova MJ, Garlan RW, DiMiceli S and Spiegel D: Psychological distress and pain significantly increase before death in metastatic breast cancer patients. Psychosom Med. 65:416–426. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Bischoff K, Weinberg V and Rabow MW: Palliative and oncologic co-management: Symptom management for outpatients with cancer. Support Care Cancer. 21:3031–3037. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Chase DM, Huang HQ, Wenzel L, Cella D, McQuellon R, Long HJ, Moore DH and Monk BJ: Quality of life and survival in advanced cervical cancer: A Gynecologic Oncology Group study. Gynecol Oncol. 125:315–319. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Kelsen DP, Portenoy RK, Thaler HT, Niedzwiecki D, Passik SD, Tao Y, Banks W, Brennan MF and Foley KM: Pain and depression in patients with newly diagnosed pancreas cancer. J Clin Oncol. 13:748–755. 1995. View Article : Google Scholar : PubMed/NCBI

8 

Turk DC, Sist TC, Okifuji A, Miner MF, Florio G, Harrison P, Massey J, Lema ML and Zevon MA: Adaptation to metastatic cancer pain, regional/local cancer pain and non-cancer pain: Role of psychological and behavioral factors. Pain. 74:247–256. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Lötsch J, Geisslinger G and Tegeder I: Genetic modulation of the pharmacological treatment of pain. Pharmacol Ther. 124:168–184. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Bell GC, Donovan KA and McLeod HL: Clinical implications of opioid pharmacogenomics in patients with cancer. Cancer Control. 22:426–432. 2015.PubMed/NCBI

11 

Rakvåg TT, Klepstad P, Baar C, Kvam TM, Dale O, Kaasa S, Krokan HE and Skorpen F: The Val158Met polymorphism of the human catechol-O-methyltransferase (COMT) gene may influence morphine requirements in cancer pain patients. Pain. 116:73–78. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Rakvåg TT, Ross JR, Sato H, Skorpen F, Kaasa S and Klepstad P: Genetic variation in the catechol-O-methyltransferase (COMT) gene and morphine requirements in cancer patients with pain. Mol Pain. 4:642008. View Article : Google Scholar : PubMed/NCBI

13 

Reyes-Gibby CC, Shete S, Rakvåg T, Bhat SV, Skorpen F, Bruera E, Kaasa S and Klepstad P: Exploring joint effects of genes and the clinical efficacy of morphine for cancer pain: OPRM1 and COMT gene. Pain. 130:25–30. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Candiotti KA, Yang Z, Buric D, Arheart K, Zhang Y, Rodriguez Y, Gitlin MC, Carvalho E, Jaraba I and Wang L: Catechol-o-methyltransferase polymorphisms predict opioid consumption in postoperative pain. Anesth Analg. 119:1194–1200. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Klepstad P, Fladvad T, Skorpen F, Bjordal K, Caraceni A, Dale O, Davies A, Kloke M, Lundström S, Maltoni M, et al: European Association for Palliative Care Research Network: Influence from genetic variability on opioid use for cancer pain: A European genetic association study of 2294 cancer pain patients. Pain. 152:1139–1145. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Atkinson TM, Mendoza TR, Sit L, Passik S, Scher HI, Cleeland C and Basch E: The Brief Pain Inventory and its ‘pain at its worst in the last 24 hours’ item: Clinical trial endpoint considerations. Pain Med. 11:337–346. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Matsuoka H, Arao T, Makimura C, Takeda M, Kiyota H, Tsurutani J, Fujita Y, Matsumoto K, Kimura H, Otsuka M, et al: Expression changes in arrestin β 1 and genetic variation in catechol-O-methyltransferase are biomarkers for the response to morphine treatment in cancer patients. Oncol Rep. 27:1393–1399. 2012.PubMed/NCBI

18 

Oken MM, Creech RH, Tormey DC, Horton J, Davis TE, McFadden ET and Carbone PP: Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 5:649–655. 1982. View Article : Google Scholar : PubMed/NCBI

19 

Swarm R, Abernethy AP, Anghelescu DL, Benedetti C, Blinderman CD, Boston B, Cleeland C, Coyle N, Deleon-Casasola OA, Eilers JG, et al: NCCN Adult Cancer Pain: Adult cancer pain. J Natl Compr Canc Netw. 8:1046–1086. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Caraceni A, Cherny N, Fainsinger R, Kaasa S, Poulain P, Radbruch L and De Conno F: Pain measurement tools and methods in clinical research in palliative care: Recommendations of an expert working group of the European Association of Palliative Care. J Pain Symptom Manage. 23:239–255. 2002. View Article : Google Scholar : PubMed/NCBI

21 

National Cancer Institute: Common Terminology Criteria for Adverse Events (CTCAE) v4.03, . https://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-14_QuickReference_5x7.pdfJune 14–2010

22 

Montagna P: Recent advances in the pharmacogenomics of pain and headache. Neurol Sci. 28 Suppl 2:S208–S212. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Farrar JT, Polomano RC, Berlin JA and Strom BL: A comparison of change in the 0–10 numeric rating scale to a pain relief scale and global medication performance scale in a short-term clinical trial of breakthrough pain intensity. Anesthesiology. 112:1464–1472. 2010. View Article : Google Scholar : PubMed/NCBI

24 

Tunbridge EM, Harrison PJ and Weinberger DR: Catechol-o-methyltransferase, cognition, and psychosis: Val158Met and beyond. Biol Psychiatry. 60:141–151. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Zubieta JK, Heitzeg MM, Smith YR, Bueller JA, Xu K, Xu Y, Koeppe RA, Stohler CS and Goldman D: COMT val158Met genotype affects mu-opioid neurotransmitter responses to a pain stressor. Science. 299:1240–1243. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Arnold BS, Alpers GW, Süss H, Friedel E, Kosmützky G, Geier A and Pauli P: Affective pain modulation in fibromyalgia, somatoform pain disorder, back pain, and healthy controls. Eur J Pain. 12:329–338. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Lee YH, Kim JH and Song GG: Association between the COMT Val158Met polymorphism and fibromyalgia susceptibility and fibromyalgia impact questionnaire score: A meta-analysis. Rheumatol Int. 35:159–166. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2017
Volume 7 Issue 4

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Matsuoka H, Makimura C, Koyama A, Fujita Y, Tsurutani J, Sakai K, Sakamoto R, Nishio K and Nakagawa K: Prospective replication study implicates the catechol-O-methyltransferase Val158Met polymorphism as a biomarker for the response to morphine in patients with cancer. Biomed Rep 7: 380-384, 2017
APA
Matsuoka, H., Makimura, C., Koyama, A., Fujita, Y., Tsurutani, J., Sakai, K. ... Nakagawa, K. (2017). Prospective replication study implicates the catechol-O-methyltransferase Val158Met polymorphism as a biomarker for the response to morphine in patients with cancer. Biomedical Reports, 7, 380-384. https://doi.org/10.3892/br.2017.963
MLA
Matsuoka, H., Makimura, C., Koyama, A., Fujita, Y., Tsurutani, J., Sakai, K., Sakamoto, R., Nishio, K., Nakagawa, K."Prospective replication study implicates the catechol-O-methyltransferase Val158Met polymorphism as a biomarker for the response to morphine in patients with cancer". Biomedical Reports 7.4 (2017): 380-384.
Chicago
Matsuoka, H., Makimura, C., Koyama, A., Fujita, Y., Tsurutani, J., Sakai, K., Sakamoto, R., Nishio, K., Nakagawa, K."Prospective replication study implicates the catechol-O-methyltransferase Val158Met polymorphism as a biomarker for the response to morphine in patients with cancer". Biomedical Reports 7, no. 4 (2017): 380-384. https://doi.org/10.3892/br.2017.963