Clinical Research INTRODUCTION. Gwieun Yeo 1, Mi Kyoung Lee 1, Heezoo Kim 1, Myounghoon Kong 1, Hyo Jung Son 2, and Han Byeol Oh 2

Similar documents
Post-operative nausea and vomiting after gynecologic laparoscopic surgery: comparison between propofol and sevoflurane

Droperidol has comparable clinical efficacy against both nausea and vomiting

Efficacy of palonosetron for the prevention of postoperative nausea and vomiting: a randomized, double-blinded, placebo-controlled trial

DEXAMETHASONE WITH EITHER GRANISETRON OR ONDANSETRON FOR POSTOPERATIVE NAUSEA AND VOMITING IN LAPAROSCOPIC SURGERY

Management of Postoperative Nausea and Vomiting in Ambulatory Surgery The Big Little Problem

4/18/2018. Disclosure. Learning Objectives. St. Vincent Healthcare. Supportive Literature. Background

IJMDS January 2017; 6(1) Dr Robina Makker Associate professor 2 Dr Amit Bhardwaj

The Journal of International Medical Research 2011; 39:

Palonosetron vs Ondansetron for prevention of postoperative nausea and vomiting in...

ISSN X (Print) India. *Corresponding author Dr. D. Shiva Prasad

Dexamethasone combined with other antiemetics for prophylaxis after laparoscopic cholecystectomy

J Med Assoc Thai 2016; 99 (5): Full text. e-journal:

Editor s key points. M. S. Green*, P. Green, S. N. Malayaman, M. Hepler, L. J. Neubert and J. C. Horrow

Incidence of and risk factors for postoperative nausea and vomiting at a Japanese Cancer Center: first large-scale study in Japan

Efficacy of a single-dose ondansetron for preventing post-operative nausea and vomiting

Alizaprideand ondansetronin the prevention of postoperative nausea and vomiting: a prospective, randomized, double-blind, placebocontrolled

Low-dose Granisetron for the Prevention of Postoperative Nausea and Vomiting

Efficacy of Prophylactic Ondansetron in a Patient-controlled Analgesia Environment

Ondansetron, a selective blocking agent of the. Timing of ondansetron administration to prevent postoperative nausea and vomiting

Satisfactory Analgesia Minimal Emesis in Day Surgeries. (SAME-Day study) A Randomized Control Trial Comparing Morphine and Hydromorphone

Dhawal R. Wadaskar*, Jyoti S. Magar, Bharati A. Tendolkar

Measure Abbreviation: PONV 01 (MIPS 430)

Measure Abbreviation: PONV 01 (MIPS 430)

Antiemetic Effect Of Propofol Administered At The End Of Surgery

Page 1 of 6 PATIENT PRESENTATION PROPHYLAXIS. No risk factors from the categories

JMSCR Vol 07 Issue 04 Page April 2019

Postoperative nausea and vomiting prophylaxis: The efficacy of a novel antiemetic drug (palonosetron) combined with dexamethasone

Setting The setting was secondary care. The economic study was carried out in the USA.

A Cross Sectional Study on Postoperative Nausea and Vomiting Risk Prediction and Assessment in Patients Undergoing Elective Surgical Patients

COMPARISON OF INDUCTION WITH SEVOFLURANE-FENTANYL AND PROPOFOL-FENTANYL ON POSTOPERATIVE NAUSEA AND VOMITING AFTER LAPAROSCOPIC SURGERY

The Comparison Effect of Preoperative Ondansetron and Metoclopramide in Reducing Nausea and Vomitting after Loparoscopic Cholecystectomy

Palonosetron versus ondansetron as rescue medication for postoperative nausea and vomiting: a randomized, multicenter, open-label study

Dexamethasone Compared with Metoclopramide in Prevention of Postoperative Nausea and Vomiting in Orthognathic Surgery

Comparison of surgical site and patient s history with a simplified risk score for the prediction of postoperative nausea and vomiting

Received: 29/05/2017 Revised: 11/09/2017 Accepted: 26/07/2017 ABSTRACT

Prevention CINV (highly emetogenic) Prevention PONV

Saeyoung Kim 1, Younghoon Jeon 2, Hyeonjun Lee 1, Jung A Lim 1, Sungsik Park 1, Si Oh Kim 1 INTRODUCTION. Original Article

Is Intravenous Ramosetron 0.3 mg Effective In The Prevention Of Postoperative Nausea And Vomiting In Women Undergoing Gynecologic Surgery?

Gastrointestinal and urinary complications in the postoperative period

Nausea and vomiting after surgery

Comparison of Drugs and Intravenous Crystalloid in Reduction of Postoperative Nausea and Vomiting after Laparoscopic Surgery

Jahan Porhomayon *, Pamela K. Wendel **, Abstract

Clinical Study Effective Dose of Ramosetron for Prophylaxis of Postoperative Nausea and Vomiting in High-Risk Patients

Pre-medication with controlled-release oxycodone in the management of postoperative pain after ambulatory laparoscopic gynaecological surgery

KEYWORDS Obstetric Patient, Spinal Anesthesia, PONV (Post-Operative Nausea and Vomiting), Inj. Metoclopramide, Inj. Ondansetron.

Comparison of Bier's Block and Systemic Analgesia for Upper Extremity Procedures: A Randomized Clinical Trial

LOW DOSE INTRAVENOUS MIDAZOLAM FOR PREVENTION OF PONV, IN LOWER ABDOMINAL SURGERY

Materials and Methods

Eun Jin Ahn, 1 Geun Joo Choi, 2 Hyun Kang, 2 Chong Wha Baek, 2 Yong Hun Jung, 2 and Young Cheol Woo Introduction. 2. Materials and Methods

A comparative study of the antiemetic efficacy of dexamethasone, ondansetron, and metoclopramide in patients undergoing gynecological surgery

IJBCP International Journal of Basic & Clinical Pharmacology

Research Article. Shital S. Ahire 1 *, Shweta Mhambrey 1, Sambharana Nayak 2. Received: 22 July 2016 Accepted: 08 August 2016

Effects of palonosetron and ondansetron on preventing nausea and vomiting after laparoscopic surgery

New Drugs in Pediatric Anesthesia

A risk score-dependent antiemetic approach effectively reduces postoperative nausea and vomiting a continuous quality improvement initiative

American Journal of Oral Medicine and Radiology

Finding the Ideal Regimen for Fentanyl-Based Intravenous Patient-Controlled Analgesia: How to Give and What to Mix?

The Incidence of Postoperative Nausea and Vomiting after Thyroidectomy using Three Anaesthetic Techniques

Won-Suk Lee 1, Kwang-Beom Lee 2, Soyi Lim 2 and Young Gin Chang 3*

Management of post-strabismus nausea and vomiting in children using ondansetron: a value-based comparison of outcomes 1^

A survey of dental treatment under general anesthesia in a Korean university hospital pediatric dental clinic

Gi-Soo Lee, Chan Kang*, You Gun Won, Byung-Hak Oh, June-Bum Jun

Factors Associated with Postoperative Nausea and Vomiting in Patients Undergoing an Ambulatory Hand Surgery

Effects of high intraoperative inspired oxygen on postoperative nausea and vomiting in gynecologic laparoscopic surgery

Reexamining Metoclopramide s Role in the Prevention of Postoperative Nausea and Vomiting: A Secondary Analysis

General anesthetics. Dr. Shamil AL-Noaimy Lecturer of Pharmacology Dept. of Pharmacology College of Medicine

Breast Augmentation: Reducing Postoperative Nausea and Vomiting. A Prospective Study

Supplemental Digital Content 1. Incidence of early nausea: Cumulative incidence 0-6h postoperatively

Postoperative Nausea and Vomiting: Update on Prevention, Rescue, and Novel Strategies

Postoperative nausea and vomiting

Volatile anaesthetics may be the main cause of early but not delayed postoperative vomiting: a randomized controlled trial of factorial design

Type of intervention Anaesthesia. Economic study type Cost-effectiveness analysis.

CONSORT 2010 checklist of information to include when reporting a randomised trial*

Risk Assessment of Postoperative Nausea and Vomiting. Christian C. Apfel, MD Norbert Roewer, MD

Nausea and Vomiting After Strabismus-Surgery: A Randomized Comparison of Isoflurane, Enflurane, Sevoflurane and Propofol

Antiemetic Prophylaxis for Office-based Surgery

NQS Domain: Patient Safety. Measure Type: Process

As laparoscopic surgeries are gaining popularity, Original Article. Maharjan SK 1, Shrestha S 2 1. Introduction

Remifentanil. Addressing the challenges of ambulatory orthopedic procedures 1-3

Effect of premedication with mirtazapine versus ondansetron on postoperative nausea and vomiting in breast surgery

A randomised, double-blind, parallel group, multicentre study to compare the tolerability, safety, and efficacy of oxycodone with morphine in

Section: Anaesthesia. Original Article INTRODUCTION

Ping-Tsung Chen, MD; Chuang-Chi Liaw, MD

Fentanyl-induced cough is a risk factor for postoperative nausea and vomiting

Evaluation of Postoperative Complications Occurring in Patients after Desflurane or Sevoflurane in Outpatient Anaesthesia: A Comparative Study

Dexamethasone Compared with Metoclopramide in Prevention of Postoperative Nausea and Vomiting in Orthognathic Surgery

Balanced Analgesia With NSAIDS and Coxibs. Raymond S. Sinatra MD, Ph.D

The Effect of Morphine on the Incidence of Postoperative Nausea and Vomiting after Strabismus Surgery with Propofol

Antiemetic and analgesic-sparing effects of diphenhydramine added to morphine intravenous patient-controlled analgesia

Statistical data preparation: management of missing values and outliers

Original Article. Abstract. Introduction. KU Tobi, CO Imarengiaye, FE Amadasun. Key words: Antiemetic, myomectomy, spinal anaesthesia

Antiemetic Prophylaxis with Ondansetron and Methylprednisolone vs Metoclopramide and Methylprednisolone in Mild and Moderately Emetogenic Chemotherapy

Automated reminders decrease postoperative nausea and vomiting incidence in a general surgical population

Updates in Chemotherapy-Induced Nausea and Vomiting (CINV) 2017

J. Basic. Appl. Sci. Res., 2(9) , , TextRoad Publication

Prophylactic use of dexamethasone in tonsillectomy among children

Postoperative nausea and vomiting after endoscopic thyroidectomy: total intravenous vs. balanced anesthesia

Factors Contributing to a Prolonged Stay After Ambulatory Surgery

Comparison of fentanyl versus fentanyl plus magnesium as post-operative epidural analgesia in orthopedic hip surgeries

Transcription:

Anesth Pain Med 2018;13:256-263 https://doi.org/10.17085/apm.2018.13.3.256 pissn 1975-5171 ㆍ eissn 2383-7977 Clinical Research Received November 17, 2017 Revised 1st, January 29, 2018 2nd, March 15, 2018 Accepted March 20, 2018 Aprepitant prophylaxis effectively reduces preventing postoperative nausea and vomiting in patients receiving opioid based intravenous patient-controlled analgesia Gwieun Yeo 1, Mi Kyoung Lee 1, Heezoo Kim 1, Myounghoon Kong 1, Hyo Jung Son 2, and Han Byeol Oh 2 1 Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, 2 Department of Anesthesia and Pain Medicine, National Police Hospital, Seoul, Korea Corresponding author Mi Kyoung Lee, M.D., Ph.D. Department of Anesthesiology and Pain Medicine, Korea University Guro Hospital, 148 Gurodong-ro, Guro-gu, Seoul 08308, Korea Tel: 82-2-2626-1437 Fax: 82-2-2626-1438 E-mail: mknim@hotmail.com ORCID http://orcid.org/0000-0002-5603-1717 Background: Aprepitant is effective in prevention of chemotherapy-induced nausea and vomiting, when administrated with other antiemetics. We compared the effectiveness of aprepitant to ondansetron for prevention of post-operative nausea and vomiting (PONV) in patients who received a patient-controlled analgesia (PCA) containing opioids. Methods: 198 patients were randomized into two groups. The treatment group was received an aprepitant, 80 mg, and the control group received a placebo. General anesthesia with inhalational anesthetics N 2 O was performed, and PCA was supplied, which contained opioids-nsaids-ondansetron. The primary end-point was the incidence of PONV for postoperative 48 hours, and the secondary end-point was the changes in the relationship between PONV incidence and risk factors. Results: PONV incidence in the treatment group was lower than in the control group (18.6% [95% CI: 10.8 26.3], 33.3% [95% CI: 23.6 43.1], respectively, P = 0.021). Relative risk of PONV in the control group was 1.80 (95% CI: 1.08 3.00, P = 0.010). PONV scores peaked at around postoperative 6 hours, then gradually decreased in the control group but not in the treatment group, which showed lower values than the control group (P = 0.001), and no changing patterns were observed (P < 0.001). Risk factors analyzed were sex, surgery type, history of motion sickness or PONV, and smoking habits. Their effects of all risk factors except sex were abolished in the treatment group. Conclusions: Prophylactic aprepitant with ondansetron was more effective than ondansetron-only regimen in preventing PONV after volatile anesthesia with opioid-containing PCA. Aprepitant abolished the effects of most of risk factors, so it could be efficacious in a high-risk PONV group. Key Words: Aprepitant, Ondansetron, Postoperative nausea and vomiting, Pre-exposure prophylaxis. INTRODUCTION Opioid-based, intravenous, patient-controlled analgesia (PCA) is a popular and effective method for postoperative pain control. However, postoperative nausea and vomiting (PONV) is one of the intractable complications of general anesthesia, and its incidence associated with opioid use reached up to 79% [1 4]. Various antiemetics in conjunction This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited. Copyright c the Korean Society of Anesthesiologists, 2018 256

Aprepitant prophylaxis for PONV with PCA have been introduced to reduce PONV; however, the result of PONV prevention has not been satisfactory [5,6]. Among these antiemetics, because of its prophylactic efficacy and its cost effectiveness, ondansetron, 5-hydroxytryptamine receptor 3 (5-HT 3 ) antagonist has already been established in various surgical procedures requiring general anesthesia. However, many clinical trials reported that ondansetron was not enough for reducing PCA related PONV [7,8]. Aprepitant is a neurokinin-1 (NK 1 ) receptor antagonist that blocks both the central and peripheral emetic stimuli of substance P [9] with a long half-life (9 to 12 hours). With combination therapy, aprepitant is approved for use and recommended for the prevention of PONV [10 13]. It is highly expected that it could cover the short action duration of ondansetron. The purpose of this study was to determine whether or not a single-dose aprepitant prophylaxis before an operation could be beneficial in reducing PONV in patients using an opioid-based intravenous PCA that contains ondansetron alone. MATERIALS AND METHODS Before the commencement of the study, appropriate hospital Institutional Review Board approval (GR08120-003) and written informed consent were obtained. We included 267 patients for eligibility. They were between the ages of 19 and 65, met criteria of the American Society of Anesthesiologists physical status classification of I or II, and were scheduled to receive elective surgery under general anesthesia, including nitrous oxide with volatile anesthesia. They all selected PCA as a postoperative pain control. We excluded patients who were pregnant, breast-feeding, or undergoing surgery with regional anesthesia or total intravenous anesthesia. Patients who needed to keep an endotracheal tube or naso-gastric tube were also excluded, as were those undergoing surgeries including eyeball, airway, middle ear, brain, and thorax. We excluded those who experienced vomiting of any organic etiology during the preoperative period or had abnormal laboratory values (alanine aminotransferase [AST] or aspartate aminotransferase [ALT] > 100 IU/L, bilirubin > 1.8 mg/ dl, creatinine > 1.6 mg/dl) and QT prolongation, atrial fibrillation, or coronary problems on electrocardiogram. Patients who received medications metabolized by cytochrom P450 3A4 (CYP3A4) were excluded. Eventually, we enrolled 198 patients to two groups randomly, using a computer-generated random number table. The patients in the treatment group (n = 103) took single-dose oral aprepitant 80mg with a small amount of clear water 1 hour before induction of anesthesia. Meanwhile, the patients in the control group (n = 95) were administrated a placebo 1 hour before induction of anesthesia. All procedures were carried out by 2 independent anesthesiologists not involved in the study. A blinded independent anesthesiologist prepared the aprepitant 80 mg and the placebo, which was the same shape as the aprepitant 80 mg capsule with a sealed opaque envelope, and another blinded independent anesthesiologist administrated to the patients. These two anesthesiologists did not participate in any remaining study process. Before the operation, we recorded patients histories, such as motion sickness, PONV, and smoking, which could affect PONV. The scheduled surgery was also recorded and subdivided into surgery, including of the abdominal area and surgery involving peripheral extremities and the abdominal area. Surgeries for fractures involving upper and lower extremities, arthroscopic surgery, arthroplasty, tendon release, and others were categorized intro surgery involving peripheral extremities and not the abdominal area. Premedication with intramuscular injection of glycopyrrolate 0.2 mg and midazolam 2 mg was done to both groups. General anesthesia was induced with thiopental (4 5 mg/kg) or propofol (1.5 2 mg/kg) and neuromuscular blocking agents. Anesthesia was maintained with nitrous oxide (40% 60%) with volatile anesthetics. And a neuromuscular block was fully reversed with intravenous pyridostigmine and glycopyrrolate prior to extubation of the trachea (train of four > 0.9). At the end of surgery, we connected PCA to the patients of two groups. The PCA regimen consisted of 700 mg fentanyl, 4 mg hydromorphone, 150 mg ketorolac, and 16 mg ondansetron. A PCA infuser (AP 0605 Ana plus, EWHA meditech, Korea) was made to deliver about 0.5 ml/hour as continuous infusion and 0.5 ml per demand with a 15-minute lockout for 48 hours period. A bolus injection to increase the opioid plasma concentration level up to adequate analgesia was prepared with the dose of 50 mg fentanyl, 0.3 mg of hydromorphone, and 10 mg ketorolac, and this was administrated within postoperative 1 hour. Investigators who were blinded to the patient s group assessed the incidence and severity of nausea, vomiting, and pain intensity at 0.5, 1, 2, 6, 12, 24, and 48 hours postop- KSAP www.anesth-pain-med.org 257

Anesth Pain Med Vol. 13 No. 3 eratively. The severity of nausea and the use of additional antiemeitics were recorded. Nausea severity scores were expressed using an 11-point verbal rating scale, with 0 meaning no nausea and 10 meaning worst possible nausea. Vomiting or retching episodes were also recorded separately. Pain intensity scores were measured on a visual analogue scale (VAS), with 0 cm meaning no pain and 10 cm meaning worst pain imaginable. Additional antiemetic therapy was given to patients who showed more than one episode of emesis at a time or nausea lasting longer than 15 minutes, and at their request. The patients could receive another pain control medication if they complained of pain 5 cm on the VAS. During the study period, the occurrence of frequently reported side effects of aprepitant, such as asthenia, fatigue, hiccups, constipation, diarrhea, and anorexia was assessed. Laboratory adverse events, such as proteinuria, increase of ALT, AST, blood urea nitrogen, and serum creatinine, were assessed using postoperative laboratory data. For blindness during the study period, all case reports were kept in closed envelopes until statistical analysis. The primary end-point was the overall incidence of the two groups during postoperative 48 hours. The secondary endpoint was the relationship changes between PONV incidence and the risk factors according to aprepitant prophylaxis. All data was expressed as mean ± standard deviation or an estimated value with a 95% confidence interval (95% CI). All variables were evaluated for normality using the Shapiro- Wilk test. Statistical analysis was performed using chi-square test for categorical variables, z-test for rate, and t-test or twoway repeated measures analysis of variance (RM-ANOVA) for numerical variables. For two-way RM-ANOVA, the sphericity assumption was evaluated with Mauchly s test, and according to those results, the Greenhouse-Geisser correction was performed. Correlation analysis was also performed to evaluate risk factors. Under 0.05 of a error probability was regarded as statistical significance. All statistical analysis was performed using IBM SPSS 23 for windows (IBM Co., USA) and Microsoft Office Excel 2010 (Microsoft Corp., USA). The study was powered to achieve at least 80%. The as- Total 267 patients were assessed for eligibility Excluded (n = 69) - Exclusion criteria (n = 57) - Patient refusal (n = 12) 198 patients were enrolled Randomized allocation Allocated to control group (n = 95) - Received allocation medication (n =94) - Did not received allocation medication* (n =1) (due to administration failure) Discontinued observation (n = 2) (due to uncovered blindness) Follow up refusal (n = 1) Analyzed (n = 90) Excluded from analysis (n = 1) (due to vague records in case report form) Allocated to treatment group (n = 103) - Received allocation medication (n =101) - Did not received allocation medication* (n =2) (due to administration failure [n = 1], inadequate administration time [n = 1]) Discontinued observation (n = 1) (due to uncovered blindness) Analyzed (n = 97) Excluded from analysis (n = 3) (due to vague records in case report form) Fig. 1. Study flowchart. *Two patients were not received aprepitant or placebo due to vomiting induced by unscheduled nasogastric tube insertion. One patient in treatment group was also excluded because the scheduled operation was delayed for few hours. They are received active aprepitant or placebo adequately, but blindness was not kept to the physician who monitored study patients during check procedure of obscure documentation about randomization number in case report form. Incorrect recording method or inaccurate writing on case report form. 258 www.anesth-pain-med.org

Aprepitant prophylaxis for PONV sumption ran as follows: 2-sided significant level of 0.05, the probability of PONV incidence of 40% in the control group and of 30% in the treatment group. Under the assumption of the c 2 distribution, the calculated effect size w was 0.204 and the required sample size was 189. We presumed that the expected exclusion rate was 30% during patient enrollment and 10% during the rest of the period of the study. As a result, the total required sample size for eligibility assessment reached 250. RESULTS Among the 267 patients who were assessed for eligibility, 69 patients were excluded due to exclusion criteria and patient refusal. In all, 198 patients were allocated into two groups according to computer generated randomization numbers. Two patients received neither the aprepitant nor the placebo, due to vomiting induced by unscheduled nasogastric tube insertion. One patient in the treatment group was also excluded because the scheduled operation was delayed for a few hours. During postoperative periods, uncovered blindness occurred in 3 patients, and one patient refused a followup visit for assessment of PONV. Penultimately, 189 patients were included in the statistical analysis. After uncovering blindness, an additional 2 patients were excluded due to vague records and missing data in case report forms. Finally, 187 patients were included in the statistical analysis (Fig. 1). There were no statistical differences between control and treatment groups in patient characteristics (Table 1). The proportions of PONV incidence varied in the two groups. Thirty patients presented PONV in the control group (n = 90) and 18 patients in treatment group (n = 97). The proportions of presenting PONV were significantly related to the groups (P = 0.032). Overall, PONV incidences during the 48 hours after surgery were significantly different between the two groups (P = 0.021). The incidence was 33.3% in the control group (95% CI: 23.6 43.1) and 18.6% in the treatment group (95% CI: 10.8 26.3). The relative risk of PONV in the control group compared to the treatment group was 1.80 and 95% CI ranging from 1.08 to 3.00 (P = 0.010). The achieved power was 0.989 under the expected incidences as above. The PONV score in the control group peaked at around 6 hours from the end time of the surgery, after which it decreased as time went by. But in the treatment group, the PONV scores during the observation periods remained at lower levels, without significant changes, even at around 6 hours from the end time of the surgery (P = 0.002). The overall PONV score in the treatment group was significantly lower than that in the control group and would have been so even if the time effect were abolished (P = 0.002, Table 2). During the 48 hours after surgery, there were no differences in pain scores between two groups (P = 0.260). The mean pain scores gradually decreased over time (P < 0.001, Table 3). The incidences of vomiting or retching were also significantly higher in the control group than in the treatment group (P = 0.001). The incidences of vomiting or retching KSAP Table 1. Demographic Data of Patients Enrolled to Study Protocol Variable Control group (n = 90) Treatment group (n = 97) Age (yr) 47.1 ± 11.3 43.9 ± 11.3 0.053 Body mass index (kg/m 2 ) 24.2 ± 3.1 24.0 ± 3.7 0.638 Duration of anesthesia (min) 143.5 ± 83.5 144.6 ± 132.8 0.780 Sex (M/F) 35/55 30/67 0.253 Surgery type (abdomen/peripheral) 57/33 49/48 0.077 History of motion sickness 33 29 0.326 History of PONV 6 9 0.511 Current smoker 15 18 0.735 Number of PONV risk factors* 0 14 24 0.408 1 42 45 2 27 22 3 6 6 4 1 0 Values are presented as mean ± SD or number of patients. PONV: post-operative nausea and vomiting. *Female; surgery type; history of motion sickness; non-smoker. www.anesth-pain-med.org 259

Anesth Pain Med Vol. 13 No. 3 during the postoperative 48 hours were 27.8% (95% CI: 18.5 37.0) in the control group and 9.3% (95% CI: 3.5 15.1) in the treatment group. The relative risk of vomiting or retching in the control group compared to the treatment group was 3.0 (95% CI: 1.5 6.1, P < 0.001). Correlation analysis revealed significant risk factors of PONV (Table 4). Following these results, we decided the risk factors of PONV were sex, surgery type, history of motion sickness or PONV, and smoking habits. Compared to the correlation coefficients of these four risk factors, aprepitant prophylaxis more strongly correlated with a reduced incidence Table 2. Overall Incidence and Verbal Rating Score of Post-operative Nausea and Vomiting (PONV) Changes during Postoperative 48 Hours Variable Control group Treatment group Overall incidence 33.3 18.6 0.021 (23.6 43.1) (10.8 26.3) Mean scores at correspondence postoperative times (h) 0.5 0.5 ± 1.4 0.4 ± 1.3 0.002* 1 0.7 ± 1.8 0.4 ± 1.3 0.002 2 1.6 ± 2.4 0.7 ± 1.8 6 1.7 ± 2.6 0.5 ± 1.4 12 1.3 ± 2.3 0.4 ± 1.4 24 0.8 ± 1.6 0.5 ± 1.6 48 0.3 ± 0.8 0.1 ± 0.4 Values are presented as percentage (95% confidence interval) or mean ± SD. *Mean PONV scores in control group were increased until postoperative 6 hours then decreased, but in treatment group, mean scores remained lower than control group (P = 0.002). Overall PONV scores in treatment group was lower than in control group (P = 0.002). of PONV. In the subgroup analysis, all 4 risk factors presented significant correlation with PONV in the control group. However, only sex significantly correlated with PONV in the treatment group (Table 4). There were no serious aprepitant-related complications during the study periods, including asthenia, fatigue, hiccups, constipation, diarrhea, and anorexia, and there were no laboratory adverse events, such as proteinuria, increase of ALT, AST, blood urea nitrogen, and serum creatinine, which were apparently related to the use of aprepitant. DISCUSSION In this randomized, double-blind, active-control case study, in our efforts to identify to what extent single-dose aprepitant prophylaxis before general anesthesia reduced PONV in patients using an opioid-based PCA containing Table 3. Visual Analogue Score of Pain Changes during Postoperative 48 Hours Postoperative time (h) Control group Treatment group 0.5 5.7 ± 2.1 6.4 ± 1.7 < 0.001* 1 5.0 ± 2.1 5.3 ± 1.8 0.260 2 4.5 ± 2.1 4.9 ± 1.8 6 3.9 ± 2.2 4.1 ± 1.9 12 3.5 ± 2.3 3.5 ± 1.7 24 2.5 ± 1.8 2.6 ± 1.7 48 1.7 ± 1.3 1.5 ± 1.1 Values are presented as mean ± SD. *Mean pain scores in both groups progressively decreased over time (P < 0.001). There was no difference in pain scores between two groups (P = 0.260). Table 4. Correlation Analysis between Post-operative Nausea and Vomiting (PONV) Incidence and Other Factors Risk factors Correlation coefficient Overall Control group Treatment group Correlation coefficient Correlation coefficient Aprepitant prophylaxis 0.308 < 0.001* N/A N/A N/A N/A Type of surgery 0.230 0.002* 0.236 0.025* 0.152 0.135 History of motion sickness or PONV 0.269 < 0.001* 0.355 0.001* 0.175 0.085 Sex 0.226 0.002* 0.257 0.014* 0.290 0.004* Age 0.031 0.676 0.031 0.774 0.032 0.756 Current smoking habit 0.161 0.028* 0.212 0.043* 0.120 0.238 Post-operative opioid usage 0.059 0.421 0.127 0.231 0.073 0.474 Maximal pain score (VAS) 0.043 0.557 0.055 0.606 0.097 0.341 Duration of surgery 0.036 0.625 0.044 0.682 0.185 0.068 Body mass index 0.025 0.739 0.036 0.738 0.016 0.878 N/A: not available, VAS: visual analogue score. *P < 0.05 with 2-tailed analysis of correlation coefficient. 260 www.anesth-pain-med.org

Aprepitant prophylaxis for PONV ondansetron, we could observe that aprepitant prophylaxis effectively reduced the incidence and severity of PONV, and aprepitant prophylaxis has a potential for modify the effect of the risk factors that were correlated with the incidence of PONV. The pathogenesis of PONV is complicated because of the involvement of multiple receptors and stimuli. Neurotransmitter receptor systems, such as dopaminergic (D 2 ), cholinergic (muscarinic), histaminergic (H 1 ), serotonergic (5- HT 3 ) and NK 1 systems mediate the signals leading to PONV [10,14,15]. Besides, PONV depends on various factors, including types of anesthesia or surgery, patient characteristics, and the contents of a PCA. The presentation of PONV may take place early, occurring up to 6 hours after the end of surgery, or late, occurring up to 24 or 48 hours after the end of surgery. The use of volatile anesthetics may be a main cause of early PONV, and opioid-induced symptoms may account for much of late PONV [6,12,16]. In this study, PONV incidence still reached up to 33% in a group supplied with only ondansetron (5-HT 3 receptor antagonist). The severity of nausea in this group increased until 6 hours postoperatively, after which it decreased gradually. This result is consistent with those of earlier studies, which reported that ondansetron alone was not enough for prevention of PONV [7,17]. Aprepitant is approved for use and recommended in consensus guidelines for the prevention of chemotherapyinduced nausea and vomiting [18]. Considering more than half of the patients who receive chemotherapy experience chemotherapy-induced nausea and vomiting, we hypothesized that a single dose of oral aprepitant prophylaxis before an operation, in conjunction with ondansetron in PCA, could be an effective regimen to alleviate PONV. Aprepitant is a highly selective, brain-penetrant NK1 receptor antagonist with a long half-life and preclinical efficacy against opioidinduced emesis [19 21]. Aprepitant can reduce the emetic effect of substance P, which is found in the gastrointestinal tract and areas of the central nervous system thought to be involved in the vomiting reflex. In our study, the patients who were administered aprepitant prophylaxis before anesthesia, in conjunction with ondansetron, showed a greatly reduced PONV incidence, at 18.6%, and a score of PONV maintained at a lower level without significant changes during the postoperative 48 hours. This is in close agreement with the results of previous studies, which reported that aprepitant was superior to ondansetron alone for the prevention of vomiting, especially in the first 24 and 48 hours postoperatively [9,11,12]. The advantages of aprepitant for PONV include its oral formulation and its easy administration for prophylaxis, along with the premedication. Also, due to the long-lasting effects of this drug, up to 24 hours, it can have advantages against PONV and efficacy against opioid-induced emesis. The reported incidence PONV associated with PCA opioids varies between 30% and 70% [1,2]. These undesirable side effects increase greatly in groups at high risk for PONV. They include female sex, surgery type, a history of motion sickness, previous PONV, nonsmoker, etc. [22,23]. It is thought that aprepitant could be beneficial in these groups of patients when they are using PCA. According to correlation analysis, type of surgery (surgery involving abdominal area), history of motion sickness, or previous PONV, female sex, current smoking habit, are the identified risk factors for PONV. However, in patients with these risk factors, aprepitant prophylaxis was most strongly and significantly correlated with PONV incidence reduction. This means that aprepitant prophylaxis could have a potential role in ameliorating the effects of other risk factors related to the occurrence of PONV. Subgroup analysis results revealed that correlation coefficients of type of surgery, history of motion sickness or PONV, and current smoking habit showed insignificant statistical results. Only sex was statistically significant. Previous trials about the effect of aprepitant in preventing PONV revealed that 40 mg aprepitant was sufficient to prevent PONV effectively [11,12]. All patients enrolled in our study were more exposed to risk factors for PONV, including potent halogenated inhalational anesthetics with nitrous oxide and continuous infusion of opioids from intravenous PCA, than those in other clinical trials. The situation is not exactly same with regard to exposure to emetogenic chemotherapy. In that case, we assumed that an opioid-containing intravenous PCA delivers similar results in mild to moderate emetogenic situations, and an 80 mg aprepitant instead of 40 mg could be better in achieving desirable results. Moreover, in our country, only 80 mg and 125 mg capsular forms of aprepitant were commercially available, so we decided to use the 80 mg single-dose prophylactic aprepitant with ondansetron for PONV prophylaxis with expectation of a nearly complete response. For using an active control-case design and avoiding ethi- KSAP www.anesth-pain-med.org 261

Anesth Pain Med Vol. 13 No. 3 cal issue, ondansetron was administered to both groups, and this was a limitation of this study. However, considering the duration or the effects of ondansetron, aprepitant prophylaxis was expected to reduce PONV by a considerable degree, compared to other risk factor modifications. Also, this study is expected to result in a more generic consequence, as it covers most eligible types of surgery that are subject to a study period and is not confined to a specific surgery type. Due to exclusion criteria, we excluded the majority of upper abdominal surgeries, which require a naso-gastric tube. Surgery involving the abdominal area mostly included lower abdominal surgery, and gynecological surgery. The peripheral surgery types were orthopedic and plastic surgery involving extremities, including arthroplasty, fracture fixation, and so on. This would not produce a homogenous result. However, we included high- and low-risk PONV surgical types, and we confirmed that the use of aprepitant prophylaxis could modify the adverse effects of most PONV risk factor, including high-risk surgery. In conclusion, a single dose of prophylactic aprepitant before an operation could be beneficial in reducing PONV in patients using opioid-based IV PCA containing ondansetron alone. The prophylactic aprepitant with ondansetron seems to be a promising regimen as a prophylactic antiemetic, even in patients with high risk factors for PONV. Its long-lasting effects, up to 48 hours, could be effective not only for early PONV but for late PONV, which is highly correlated with the use of opioids for postoperative pain control. REFERENCES 1. Hong D, Flood P, Diaz G. The side effects of morphine and hydromorphone patient-controlled analgesia. Anesth Analg 2008; 107: 1384-9. 2. Kim SY, Kim EM, Nam KH, Chang DJ, Nam SH, Kim KJ. Postoperative intravenous patient-controlled analgesia in thyroid surgery: comparison of fentanyl and ondansetron regimens with and without the nonsteriodal anti-inflammatory drug ketorolac. Thyroid 2008; 18: 1285-90. 3. Apfel CC, Greim CA, Haubitz I, Goepfert C, Usadel J, Sefrin P, et al. A risk score to predict the probability of postoperative vomiting in adults. Acta Anaesthesiol Scand 1998; 42: 495-501. 4. Apfel CC, Greim CA, Haubitz I, Grundt D, Goepfert C, Sefrin P, et al. The discriminating power of a risk score for postoperative vomiting in adults undergoing various types of surgery. Acta Anaesthesiol Scand 1998; 42: 502-9. 5. Kwak KH. PONV prevention: still not enough. Korean J Anesthesiol 2017; 70: 489-90 6. Apfel CC, Korttila K, Abdalla M, Kerger H, Turan A, Vedder I, et al. A factorial trial of six interventions for the prevention of postoperative nausea and vomiting. N Engl J Med 2004; 350: 2441-51. 7. Cherian VT, Smith I. Prophylactic ondansetron does not improve patient satisfaction in women using PCA after caesarean section. Br J Anaesth 2001; 87: 502-4. 8. Gan TJ, Meyer TA, Apfel CC, Chung F, Davis PJ, Habib AS, et al. Society for ambulatory anesthesia guidelines for the management of postoperative nausea and vomiting. Anesth Analg 2007; 105: 1615-28. 9. Gesztesi Z, Scuderi PE, White PF, Wright W, Wender RH, D Angelo R, et al. Substance P (neurokinin-1) antagonist prevents postoperative vomiting after abdominal hysterectomy procedures. Anesthesiology 2000; 93: 931-7. 10. Curran MP, Robinson DM. Aprepitant: a review of its use in the prevention of nausea and vomiting. Drugs 2009; 69: 1853-78. 11. Diemunsch P, Gan TJ, Philip BK, Girao MJ, Eberhart L, Irwin MG, et al. Single-dose aprepitant vs ondansetron for the prevention of postoperative nausea and vomiting: a randomized, double-blind phase III trial in patients undergoing open abdominal surgery. Br J Anaesth 2007; 99: 202-11. 12. Diemunsch P, Schoeffler P, Bryssine B, Cheli-Muller LE, Lees J, McQuade BA, et al. Antiemetic activity of the NK1 receptor antagonist GR205171 in the treatment of established postoperative nausea and vomiting after major gynaecological surgery. Br J Anaesth 1999; 82: 274-6. 13. Moon YE. Postoperative nausea and vomiting. Korean J Anesthesiol 2014; 67: 164-70. 14. Watcha MF, White PF. Postoperative nausea and vomiting. Its etiology, treatment, and prevention. Anesthesiology 1992; 77: 162-84. 15. Navari RM. Antiemetic control: toward a new standard of care for emetogenic chemotherapy. Expert Opin Pharmacother 2009; 10: 629-44. 16. Gralla RJ, Roila F, Tonato M. The 2004 Perugia antiemetic consensus guideline process: methods, procedures, and participants. Support Care Cancer 2005; 13: 77-9. 17. Hwang HY, Kim EJ, Yang MK, Kim JA, Kang JG, Kim TH, et al. The effect of multimodal approach with combination of antiemetics and total intravenous anesthesia on postoperative nausea and vomiting in very high-risk patients. Anesth Pain Med 2010; 5: 45-9. 18. Gralla RJ, Roila F, Tonato M. The 2004 Perugia antiemetic con- 262 www.anesth-pain-med.org

Aprepitant prophylaxis for PONV sensus guideline process: methods, procedures, and participants. Support Care Cancer 2005; 13: 77-9. 19. Gregory RE, Ettinger DS. 5-HT3 receptor antagonists for the prevention of chemotherapy-induced nausea and vomiting. A comparison of their pharmacology and clinical efficacy. Drugs 1998; 55: 173-89. 20. Kovac AL. Prevention and treatment of postoperative nausea and vomiting. Drugs 2000; 59: 213-43. 21. Kazemi-Kjellberg F, Henzi I, Tramèr MR. Treatment of established postoperative nausea and vomiting: a quantitative systematic review. BMC Anesthesiol 2001; 1: 2. 22. Koivuranta M, Läärä E, Snåre L, Alahuhta S. A survey of postoperative nausea and vomiting. Anaesthesia 1997; 52: 443-9. 23. Apfel CC, Läärä E, Koivuranta M, Greim CA, Roewer N. A simplified risk score for predicting postoperative nausea and vomiting: conclusions from cross-validations between two centers. Anesthesiology 1999; 91: 693-700. KSAP www.anesth-pain-med.org 263